Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 21
1.
Nat Neurosci ; 2024 May 21.
Article En | MEDLINE | ID: mdl-38773348

Retrotransposons are mobile DNA sequences duplicated via transcription and reverse transcription of an RNA intermediate. Cis-regulatory elements encoded by retrotransposons can also promote the transcription of adjacent genes. Somatic LINE-1 (L1) retrotransposon insertions have been detected in mammalian neurons. It is, however, unclear whether L1 sequences are mobile in only some neuronal lineages or therein promote neurodevelopmental gene expression. Here we report programmed L1 activation by SOX6, a transcription factor critical for parvalbumin (PV) interneuron development. Mouse PV interneurons permit L1 mobilization in vitro and in vivo, harbor unmethylated L1 promoters and express full-length L1 mRNAs and proteins. Using nanopore long-read sequencing, we identify unmethylated L1s proximal to PV interneuron genes, including a novel L1 promoter-driven Caps2 transcript isoform that enhances neuron morphological complexity in vitro. These data highlight the contribution made by L1 cis-regulatory elements to PV interneuron development and transcriptome diversity, uncovered due to L1 mobility in this milieu.

2.
Development ; 151(1)2024 Jan 01.
Article En | MEDLINE | ID: mdl-38063486

Cholinergic signaling plays a crucial role in the regulation of adult hippocampal neurogenesis; however, the mechanisms by which acetylcholine mediates neurogenic effects are not completely understood. Here, we report the expression of muscarinic acetylcholine receptor subtype M4 (M4 mAChR) on a subpopulation of neural precursor cells (NPCs) in the adult mouse hippocampus, and demonstrate that its pharmacological stimulation promotes their proliferation, thereby enhancing the production of new neurons in vivo. Using a targeted ablation approach, we also show that medial septum (MS) and the diagonal band of Broca (DBB) cholinergic neurons support both the survival and morphological maturation of adult-born neurons in the mouse hippocampus. Although the systemic administration of an M4-selective allosteric potentiator fails to fully rescue the MS/DBB cholinergic lesion-induced decrease in hippocampal neurogenesis, it further exacerbates the impairment in the morphological maturation of adult-born neurons. Collectively, these findings reveal stage-specific roles of M4 mAChRs in regulating adult hippocampal neurogenesis, uncoupling their positive role in enhancing the production of new neurons from the M4-induced inhibition of their morphological maturation, at least in the context of cholinergic signaling dysfunction.


Neural Stem Cells , Receptor, Muscarinic M4 , Mice , Animals , Receptor, Muscarinic M4/metabolism , Neural Stem Cells/metabolism , Hippocampus/metabolism , Neurogenesis/genetics , Cholinergic Agents/metabolism , Cholinergic Agents/pharmacology , Cell Proliferation
3.
eNeuro ; 10(11)2023 11.
Article En | MEDLINE | ID: mdl-37923391

Stress has been identified as a major contributor to human disease and is postulated to play a substantial role in epileptogenesis. In a significant proportion of individuals with epilepsy, sensitivity to stressful events contributes to dynamic symptomatic burden, notably seizure occurrence and frequency, and presence and severity of psychiatric comorbidities [anxiety, depression, posttraumatic stress disorder (PTSD)]. Here, we review this complex relationship between stress and epilepsy using clinical data and highlight key neurobiological mechanisms including the hypothalamic-pituitary-adrenal (HPA) axis dysfunction, altered neuroplasticity within limbic system structures, and alterations in neurochemical pathways such as brain-derived neurotrophic factor (BNDF) linking epilepsy and stress. We discuss current clinical management approaches of stress that help optimize seizure control and prevention, as well as psychiatric comorbidities associated with epilepsy. We propose that various shared mechanisms of stress and epilepsy present multiple avenues for the development of new symptomatic and preventative treatments, including disease modifying therapies aimed at reducing epileptogenesis. This would require close collaborations between clinicians and basic scientists to integrate data across multiple scales, from genetics to systems biology, from clinical observations to fundamental mechanistic insights. In future, advances in machine learning approaches and neuromodulation strategies will enable personalized and targeted interventions to manage and ultimately treat stress-related epileptogenesis.


Epilepsy , Stress Disorders, Post-Traumatic , Humans , Epilepsy/therapy , Epilepsy/complications , Seizures/complications , Stress Disorders, Post-Traumatic/psychology , Anxiety Disorders , Anxiety
4.
J Neurosci ; 43(30): 5448-5457, 2023 07 26.
Article En | MEDLINE | ID: mdl-37419688

Activity-dependent changes in the number of AMPA-type glutamate receptors (AMPARs) at the synapse underpin the expression of LTP and LTD, cellular correlates of learning and memory. Post-translational ubiquitination has emerged as a key regulator of the trafficking and surface expression of AMPARs, with ubiquitination of the GluA1 subunit at Lys-868 controlling the post-endocytic sorting of the receptors into the late endosome for degradation, thereby regulating their stability at synapses. However, the physiological significance of GluA1 ubiquitination remains unknown. In this study, we generated mice with a knock-in mutation in the major GluA1 ubiquitination site (K868R) to investigate the role of GluA1 ubiquitination in synaptic plasticity, learning, and memory. Our results reveal that these male mice have normal basal synaptic transmission but exhibit enhanced LTP and deficits in LTD. They also display deficits in short-term spatial memory and cognitive flexibility. These findings underscore the critical roles of GluA1 ubiquitination in bidirectional synaptic plasticity and cognition in male mice.SIGNIFICANCE STATEMENT Subcellular targeting and membrane trafficking determine the precise number of AMPA-type glutamate receptors at synapses, processes that are essential for synaptic plasticity, learning, and memory. Post-translational ubiquitination of the GluA1 subunit marks AMPARs for degradation, but its functional role in vivo remains unknown. Here we demonstrate that the GluA1 ubiquitin-deficient mice exhibit an altered threshold for synaptic plasticity accompanied by deficits in short-term memory and cognitive flexibility. Our findings suggest that activity-dependent ubiquitination of GluA1 fine-tunes the optimal number of synaptic AMPARs required for bidirectional synaptic plasticity and cognition in male mice. Given that increases in amyloid-ß cause excessive ubiquitination of GluA1, inhibiting that GluA1 ubiquitination may have the potential to ameliorate amyloid-ß-induced synaptic depression in Alzheimer's disease.


Neuronal Plasticity , Receptors, AMPA , Mice , Male , Animals , Receptors, AMPA/metabolism , alpha-Amino-3-hydroxy-5-methyl-4-isoxazolepropionic Acid/metabolism , Neuronal Plasticity/physiology , Synapses/physiology , Receptors, Glutamate/metabolism , Ubiquitination , Cognition , Hippocampus/metabolism
5.
NPJ Sci Learn ; 7(1): 16, 2022 Jul 16.
Article En | MEDLINE | ID: mdl-35842419

Adult hippocampal neurogenesis in the developmental process of generating and integrating new neurons in the hippocampus during adulthood and is a unique form of structural plasticity with enormous potential to modulate neural circuit function and behaviour. Dysregulation of this process is strongly linked to stress-related neuropsychiatric conditions such as anxiety and depression, and efforts have focused on unravelling the contribution of adult-born neurons in regulating stress response and recovery. Chronic stress has been shown to impair this process, whereas treatment with clinical antidepressants was found to enhance the production of new neurons in the hippocampus. However, the precise role of adult hippocampal neurogenesis in mediating the behavioural response to chronic stress is not clear and whether these adult-born neurons buffer or increase susceptibility to stress-induced mood-related maladaptation remains one of the controversial issues. In this review, we appraise evidence probing the causal role of adult hippocampal neurogenesis in the regulation of emotional behaviour in rodents. We find that the relationship between adult-born hippocampal neurons and stress-related mood disorders is not linear, and that simple subtraction or addition of these neurons alone is not sufficient to lead to anxiety/depression or have antidepressant-like effects. We propose that future studies examining how stress affects unique properties of adult-born neurons, such as the excitability and the pattern of connectivity during their critical period of maturation will provide a deeper understanding of the mechanisms by which these neurons contribute to functional outcomes in stress-related mood disorders.

6.
Neuron ; 109(10): 1593-1595, 2021 05 19.
Article En | MEDLINE | ID: mdl-34015266

The mechanism by which antidepressants elicit clinical improvements has proven elusive. In a recent publication in Cell, Casarotto et al. (2021) reveal a surprising direct interaction between antidepressants and TrkB. This link provides an important mechanistic insight into synaptic remodeling that may assist in the design of improved antidepressant therapeutics.


Nerve Growth Factors , Receptor, trkB , Antidepressive Agents/pharmacology , Protein Binding , Receptor, trkB/metabolism , Signal Transduction/drug effects
7.
Int J Biochem Cell Biol ; 134: 105969, 2021 05.
Article En | MEDLINE | ID: mdl-33727042

The production and circuit integration of new neurons is one of the defining features of the adult mammalian hippocampus. A wealth of evidence has established that adult hippocampal neurogenesis is exquisitely sensitive to neuronal activity-mediated regulation. How these signals are interpreted and contribute to neurogenesis and hippocampal functions has been a subject of immense interest. In particular, neurotransmitters, in addition to their synaptic roles, have been shown to offer important trophic support. Amongst these, acetylcholine, which has a prominent role in cognition, has been implicated in regulating neurogenesis. In this review, we appraise the evidence linking the contribution of cholinergic signalling to the regulation of adult hippocampal neurogenesis and hippocampus-dependent functions. We discuss open questions that need to be addressed to gain a deeper mechanistic understanding of the role and translational potential of acetylcholine and its receptors in regulating this form of cellular neuroplasticity.


Cholinergic Agents/pharmacology , Cognition/physiology , Hippocampus/physiology , Neurons/physiology , Adult , Animals , Cognition/drug effects , Hippocampus/drug effects , Humans , Neurogenesis , Neuronal Plasticity , Neurons/drug effects
8.
Dev Neurobiol ; 79(9-10): 868-879, 2019 09.
Article En | MEDLINE | ID: mdl-31886631

Brain-derived neurotrophic factor (BDNF) signaling plays a major role in the regulation of hippocampal neurogenesis in the adult brain. While the majority of studies suggest that this is due to its effect on the survival and differentiation of newborn neurons, it remains unclear whether this signaling directly regulates neural precursor cell (NPC) activity and which of its two receptors, TrkB or the p75 neurotrophin receptor (p75NTR ) mediates this effect. Here, we examined both the RNA and protein expression of these receptors and found that TrkB but not p75NTR receptors are expressed by hippocampal NPCs in the adult mouse brain. Using a clonal neurosphere assay, we demonstrate that pharmacological blockade of TrkB receptors directly activates a distinct subpopulation of NPCs. Moreover, we show that administration of ANA-12, a TrkB-selective antagonist, in vivo either by systemic intraperitoneal injection or by direct infusion within the hippocampus leads to an increase in the production of new neurons. In contrast, we found that NPC-specific knockout of p75NTR had no effect on the proliferation of NPCs and did not alter neurogenesis in the adult hippocampus. Collectively, these results demonstrate a novel role of TrkB receptors in directly regulating the activity of a subset of hippocampal NPCs and suggest that the transient blockade of these receptors could be used to enhance adult hippocampal neurogenesis.


Brain/metabolism , Hippocampus/metabolism , Neurogenesis/physiology , Receptor, Nerve Growth Factor/metabolism , Animals , Cell Differentiation/physiology , Cells, Cultured , Female , Male , Mice , Neural Stem Cells/metabolism , Neurons/metabolism , Signal Transduction/physiology
9.
Cereb Cortex ; 29(10): 4381-4397, 2019 09 13.
Article En | MEDLINE | ID: mdl-30590507

The hippocampal dentate gyrus (DG) is a major region of the adult rodent brain in which neurogenesis occurs throughout life. The EphA4 receptor, which regulates neurogenesis and boundary formation in the developing brain, is also expressed in the adult DG, but whether it regulates adult hippocampal neurogenesis is not known. Here, we show that, in the adult mouse brain, EphA4 inhibits hippocampal precursor cell proliferation but does not affect precursor differentiation or survival. Genetic deletion or pharmacological inhibition of EphA4 significantly increased hippocampal precursor proliferation in vivo and in vitro, by blocking EphA4 forward signaling. EphA4 was expressed by mature hippocampal DG neurons but not neural precursor cells, and an EphA4 antagonist, EphA4-Fc, did not activate clonal cultures of precursors until they were co-cultured with non-precursor cells, indicating an indirect effect of EphA4 on the regulation of precursor activity. Supplementation with d-serine blocked the increased precursor proliferation induced by EphA4 inhibition, whereas blocking the interaction between d-serine and N-methyl-d-aspartate receptors (NMDARs) promoted precursor activity, even at the clonal level. Collectively, these findings demonstrate that EphA4 indirectly regulates adult hippocampal precursor proliferation and thus plays a role in neurogenesis via d-serine-regulated NMDAR signaling.


Dentate Gyrus/metabolism , Neural Stem Cells/metabolism , Neurogenesis , Receptor, EphA4/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , Animals , Cell Differentiation , Cell Proliferation , Cell Survival , Female , Male , Mice, Inbred C57BL , Mice, Knockout , Receptor, EphA4/genetics , Signal Transduction
10.
Front Psychiatry ; 10: 974, 2019.
Article En | MEDLINE | ID: mdl-32038323

Major depressive disorder (MDD) is a global problem for which current pharmacotherapies are not completely effective. Hypothalamic-pituitary-adrenal (HPA) axis dysfunction has long been associated with MDD; however, the value of assessing cortisol as a biological benchmark of the pathophysiology or treatment of MDD is still debated. In this review, we critically evaluate the relationship between HPA axis dysfunction and cortisol level in relation to MDD subtype, stress, gender and treatment regime, as well as in rodent models. We find that an elevated cortisol response to stress is associated with acute and severe, but not mild or atypical, forms of MDD. Furthermore, the increased incidence of MDD in females is associated with greater cortisol response variability rather than higher baseline levels of cortisol. Despite almost all current MDD treatments influencing cortisol levels, we could find no convincing relationship between cortisol level and therapeutic response in either a clinical or preclinical setting. Thus, we argue that the absolute level of cortisol is unreliable for predicting the efficacy of antidepressant treatment. We propose that future preclinical models should reliably produce exaggerated HPA axis responses to acute or chronic stress a priori, which may, or may not, alter baseline cortisol levels, while also modelling the core symptoms of MDD that can be targeted for reversal. Combining genetic and environmental risk factors in such a model, together with the interrogation of the resultant molecular, cellular, and behavioral changes, promises a new mechanistic understanding of MDD and focused therapeutic strategies.

11.
Dev Dyn ; 247(1): 194-200, 2018 01.
Article En | MEDLINE | ID: mdl-28685906

BACKGROUND: Type 1 adult hippocampal neural stem cells (AH-NSCs) continue to generate neurons throughout life, albeit at a very low rate. The relative quiescence of this population of cells has led to many studies investigating factors that may increase their division. Current methods of identifying dividing AH-NSCs in vivo require the identification and tracing of radial processes back to nuclei within the subgranular zone. However, caveats to this approach include the time-intensive nature of identifying AH-NSCs with such a process, as well as the fact that this approach ignores the relatively more active population of horizontally oriented AH-NSCs that also reside in the subgranular zone. RESULTS: Here we describe, and then verify using Hes5::GFP mice, that labeling for the cell cycle marker Ki67 and selection against the intermediate progenitor cell marker TBR2 (Ki67+ve ; TBR2-ve nuclei) is sufficient to identify dividing horizontally and radially oriented AH-NSCs in the adult mouse hippocampus. CONCLUSIONS: These findings provide a simple and accurate way to quantify dividing AH-NSCs in vivo using a morphology-independent approach that will facilitate studies into neurogenesis within the hippocampal stem cell niche of the adult brain. Developmental Dynamics 247:194-200, 2018. © 2017 Wiley Periodicals, Inc.


Adult Stem Cells/cytology , Hippocampus/cytology , Microscopy, Confocal/methods , Microscopy, Fluorescence/methods , Neural Stem Cells/cytology , Neurogenesis/physiology , Animals , Cell Proliferation/physiology , Mice
12.
Psychoneuroendocrinology ; 86: 53-63, 2017 Dec.
Article En | MEDLINE | ID: mdl-28915381

Vitamin D deficiency is prevalent in adults throughout the world. Epidemiological studies have shown significant associations between vitamin D deficiency and an increased risk of various neuropsychiatric and neurodegenerative disorders, such as schizophrenia, depression, Alzheimer's disease and cognitive impairment. However, studies based on observational epidemiology cannot address questions of causality; they cannot determine if vitamin D deficiency is a causal factor leading to the adverse health outcome. The main aim of this study was to determine if AVD deficiency would exacerbate the effects of a secondary exposure, in this case social stress, in BALB/c mice and in the more resilient C57BL/6 mice. Ten-week old male BALB/c and C57BL/6 mice were fed a control or vitamin D deficient diet for 10 weeks, and the mice were further separated into one of two groups for social treatment, either Separated (SEP) or Social Defeat (DEF). SEP mice were placed two per cage with a perforated Plexiglas divider, whereas the DEF mice underwent 10days of social defeat prior to behavioural testing. We found that AVD-deficient mice were more vulnerable to the effects of social stress using a social avoidance test, and this was dependent on strain. These results support the hypothesis that vitamin D deficiency may exacerbate behavioural outcomes in mice vulnerable to stress, a finding that can help guide future studies. Importantly, these discoveries support the epidemiological link between vitamin D deficiency and neuropsychiatric and neurodegenerative disorders; and has provided clues that can guide future studies related to unravelling the mechanisms of action linking adult vitamin D deficiency and adverse brain related outcomes.


Stress, Psychological/metabolism , Vitamin D Deficiency/metabolism , Vitamin D Deficiency/psychology , Animals , Behavior, Animal/drug effects , Diet , Disease Models, Animal , Male , Mental Health , Mice , Mice, Inbred BALB C/metabolism , Mice, Inbred C57BL/metabolism , Mice, Inbred Strains , Social Behavior , Vitamin D/metabolism
13.
J Neurosci ; 35(21): 8132-44, 2015 May 27.
Article En | MEDLINE | ID: mdl-26019330

The activity of neural precursor cells in the adult hippocampus is regulated by various stimuli; however, whether these stimuli regulate the same or different precursor populations remains unknown. Here, we developed a novel cell-sorting protocol that allows the purification to homogeneity of neurosphere-forming neural precursors from the adult mouse hippocampus and examined the responsiveness of individual precursors to various stimuli using a clonal assay. We show that within the Hes5-GFP(+)/Nestin-GFP(+)/EGFR(+) cell population, which comprises the majority of neurosphere-forming precursors, there are two distinct subpopulations of quiescent precursor cells, one directly activated by high-KCl depolarization, and the other activated by norepinephrine (NE). We then demonstrate that these two populations are differentially distributed along the septotemporal axis of the hippocampus, and show that the NE-responsive precursors are selectively regulated by GABA, whereas the KCl-responsive precursors are selectively modulated by corticosterone. Finally, based on RNAseq analysis by deep sequencing, we show that the progeny generated by activating NE-responsive versus KCl-responsive quiescent precursors are molecularly different. These results demonstrate that the adult hippocampus contains phenotypically similar but stimulus-specific populations of quiescent precursors, which may give rise to neural progeny with different functional capacity.


Cell Separation , Hippocampus/cytology , Hippocampus/growth & development , Neural Stem Cells/physiology , Neurogenesis/physiology , Age Factors , Animals , Cell Count/methods , Cell Separation/methods , Cells, Cultured , Male , Mice , Mice, Inbred C57BL
14.
PLoS One ; 9(8): e103207, 2014.
Article En | MEDLINE | ID: mdl-25119138

Recent advances in next-generation sequencing technology allow high-throughput cDNA sequencing (RNA-Seq) to be widely applied in transcriptomic studies, in particular for detecting differentially expressed genes between groups. Many software packages have been developed for the identification of differentially expressed genes (DEGs) between treatment groups based on RNA-Seq data. However, there is a lack of consensus on how to approach an optimal study design and choice of suitable software for the analysis. In this comparative study we evaluate the performance of three of the most frequently used software tools: Cufflinks-Cuffdiff2, DESeq and edgeR. A number of important parameters of RNA-Seq technology were taken into consideration, including the number of replicates, sequencing depth, and balanced vs. unbalanced sequencing depth within and between groups. We benchmarked results relative to sets of DEGs identified through either quantitative RT-PCR or microarray. We observed that edgeR performs slightly better than DESeq and Cuffdiff2 in terms of the ability to uncover true positives. Overall, DESeq or taking the intersection of DEGs from two or more tools is recommended if the number of false positives is a major concern in the study. In other circumstances, edgeR is slightly preferable for differential expression analysis at the expense of potentially introducing more false positives.


Gene Expression Profiling/methods , Software , Animals , Benchmarking , Cell Line , DNA, Complementary/chemistry , High-Throughput Nucleotide Sequencing , Humans , Male , Mice, Inbred C57BL , RNA/chemistry , Reverse Transcriptase Polymerase Chain Reaction , Sequence Analysis, RNA/methods
15.
PLoS One ; 9(6): e98736, 2014.
Article En | MEDLINE | ID: mdl-24922313

Norepinephrine regulates latent neural stem cell activity and adult hippocampal neurogenesis, and has an important role in modulating hippocampal functions such as learning, memory and mood. Adult hippocampal neurogenesis is a multi-stage process, spanning from the activation and proliferation of hippocampal stem cells, to their differentiation into neurons. However, the stage-specific effects of noradrenergic receptors in regulating adult hippocampal neurogenesis remain poorly understood. In this study, we used transgenic Nestin-GFP mice and neurosphere assays to show that modulation of α2- and ß-adrenergic receptor activity directly affects Nestin-GFP/GFAP-positive precursor cell population albeit in an opposing fashion. While selective stimulation of α2-adrenergic receptors decreases precursor cell activation, proliferation and immature neuron number, stimulation of ß-adrenergic receptors activates the quiescent precursor pool and enhances their proliferation in the adult hippocampus. Furthermore, our data indicate no major role for α1-adrenergic receptors, as we did not observe any change in either the activation and proliferation of hippocampal precursors following selective stimulation or blockade of α1-adrenergic receptors. Taken together, our data suggest that under physiological as well as under conditions that lead to enhanced norepinephrine release, the balance between α2- and ß-adrenergic receptor activity regulates precursor cell activity and hippocampal neurogenesis.


Adrenergic alpha-2 Receptor Agonists/pharmacology , Adrenergic beta-Agonists/pharmacology , Hippocampus/drug effects , Neural Stem Cells/drug effects , Neurogenesis , Adrenergic alpha-2 Receptor Antagonists/pharmacology , Adrenergic beta-Antagonists/pharmacology , Animals , Cell Proliferation , Cells, Cultured , Hippocampus/cytology , Hippocampus/growth & development , Male , Mice , Mice, Inbred C57BL , Nestin/genetics , Nestin/metabolism , Neural Stem Cells/cytology , Neural Stem Cells/metabolism
16.
Methods Mol Biol ; 1059: 41-51, 2013.
Article En | MEDLINE | ID: mdl-23934832

It is now well established that a resident population of neural precursor cells continues to generate new neurons in the adult hippocampus throughout life. Numerous studies have suggested that these newborn neurons preferentially participate in the functional hippocampal circuitry that leads to enhancement of learning, cognition and mood. Therefore, understanding the molecular mechanisms that regulate the activity of these endogenous precursor cells is paramount to develop novel regenerative strategies for the treatment of neurological and psychiatric disorders. The neurosphere assay has been instrumental in discovering the presence of stem and precursor cell population from several brain regions. In this chapter, we describe this assay to specifically isolate and culture neural stem and precursor cell populations from the adult hippocampus of mice. In addition, we provide methods to conduct detailed assays to examine their functional properties such as proliferation, self-renewal, and differentiation.


Hippocampus/cytology , Neural Stem Cells/physiology , Primary Cell Culture , Spheroids, Cellular/physiology , Animals , Cell Differentiation , Cell Proliferation , Cell Separation , Culture Media , Dissection , Female , Immunohistochemistry , Male , Mice
17.
Front Cell Neurosci ; 7: 5, 2013.
Article En | MEDLINE | ID: mdl-23372544

While it is now well-established that resident populations of stem and progenitor cells drive neurogenesis in the adult brain, a growing body of evidence indicates that these new neurons play a pivotal role in spatial learning, memory, and mood regulation. As such, interest is gathering to develop strategies to harness the brain's endogenous reservoir of stem and progenitor cells, with the view that newborn neurons may help overcome the loss of neural and cognitive function that occurs during neurodegenerative disease and psychiatric illness. Here we review evidence for the presence of endogenous stem cell populations in the adult hippocampus, especially large pools of latent stem and precursor cells, and the ways in which these populations can be stimulated to produce new neurons. While the translation of this research from animal models to human application is still in its infancy, understanding in detail the cellular and molecular mechanisms that regulate endogenous neurogenesis, offers the potential to use this innate reservoir of precursors to produce neurons that may be able to mitigate against cognitive decline and mood disorders.

18.
J Neurosci Res ; 91(5): 642-59, 2013 May.
Article En | MEDLINE | ID: mdl-23404532

Within the two neurogenic niches of the adult mammalian brain, i.e., the subventricular zone lining the lateral ventricle and the subgranular zone of the hippocampus, there exist distinct populations of proliferating neural precursor cells that differentiate to generate new neurons. Numerous studies have suggested that epigenetic regulation by histone-modifying proteins is important in guiding precursor differentiation during development; however, the role of these proteins in regulating neural precursor activity in the adult neurogenic niches remains poorly understood. Here we examine the role of an NAD(+) -dependent histone deacetylase, SIRT1, in modulating the neurogenic potential of neural precursors in the neurogenic niches of the adult mouse brain. We show that SIRT1 is expressed by proliferating adult subventricular zone and hippocampal neural precursors, although its transcript and protein levels are dramatically reduced during neural precursor differentiation. Utilizing a lentiviral-mediated delivery strategy, we demonstrate that abrogation of SIRT1 signaling by RNAi does not affect neural precursor numbers or their proliferation. However, SIRT1 knock down results in a significant increase in neuronal production in both the subventricular zone and the hippocampus. In contrast, enhancing SIRT1 signaling either through lentiviral-mediated SIRT1 overexpression or through use of the SIRT1 chemical activator Resveratrol prevents adult neural precursors from differentiating into neurons. Importantly, knock down of SIRT1 in hippocampal precursors in vivo, either through RNAi or through genetic ablation, promotes their neurogenic potential. These findings highlight SIRT1 signaling as a negative regulator of neuronal differentiation of adult subventricular zone and hippocampal neural precursors. © 2013 Wiley Periodicals, Inc.


Cerebral Ventricles/cytology , Hippocampus/cytology , Neural Stem Cells/physiology , Neurogenesis/genetics , Sirtuin 1/metabolism , Analysis of Variance , Animals , Bromodeoxyuridine/metabolism , Cell Differentiation/drug effects , Cell Differentiation/genetics , Cell Proliferation/drug effects , Cells, Cultured , Dose-Response Relationship, Drug , Doublecortin Domain Proteins , Enzyme Inhibitors/pharmacology , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Ki-67 Antigen/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Microtubule-Associated Proteins/metabolism , Neurogenesis/drug effects , Neuropeptides/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Resveratrol , Sirtuin 1/deficiency , Sirtuin 1/genetics , Stem Cell Niche/genetics , Stem Cell Niche/physiology , Stilbenes/pharmacology , Transfection , Tubulin/metabolism
19.
Dev Neurobiol ; 72(7): 1044-58, 2012 Jul.
Article En | MEDLINE | ID: mdl-22505142

Resident populations of stem and precursor cells drive the production of new neurons in the adult hippocampus. Recent discoveries have highlighted that a large proportion of these precursor cells are in fact quiescent and can be activated by distinct neuronal activity under both normal physiological and pathological conditions. As growing evidence indicates that newborn neurons play a critical role in cognitive functions such as learning and memory and in mood regulation, it is paramount that we obtain a better understanding of how the reservoirs of stem and precursor cells are maintained and activated. In this review, we critically examine the roles of key molecular mechanisms that have been shown to regulate hippocampal precursor cells, especially their activation. We believe that understanding the mechanistic details of the activity-driven regulation of precursor cells will equip us with the ability to develop tailored strategies to trigger the generation of new neurons, thereby improving the functional outcomes in various neurological and psychiatric conditions.


Hippocampus/cytology , Neural Stem Cells/cytology , Neurogenesis/physiology , Neurons/cytology , Animals , Hippocampus/physiology , Learning/physiology , Neural Stem Cells/physiology , Neuronal Plasticity/physiology , Neurons/physiology
20.
Dev Neurobiol ; 71(7): 619-33, 2011 Jul.
Article En | MEDLINE | ID: mdl-21671408

The regulation of neural precursor cell (NPC) activity is the major determinant of the rate of neuronal production in neurogenic regions of the adult brain. Here, we show that Oncostatin M (Osm) and its receptor, OsmRß, are both expressed in the subventricular zone (SVZ) and that in contradistinction to leukemia inhibitory factor and ciliary neutrophic factor, Osm directly inhibits the proliferation of adult NPCs as measured by a decreased level of neurosphere formation in vitro. Similarly, intraventricular infusion of Osm dramatically decreases the pool of NPCs in both the SVZ and the hippocampus. In keeping with the inhibitory action of Osm, we reveal that mice lacking OsmRß have substantially more NPCs in the SVZ, the hippocampus and the olfactory bulb, demonstrating that endogenous Osm signaling is important for NPC homeostasis. Finally, we show that Osm can also inhibit clonal growth of glioblastoma-derived neurospheres, further supporting the close link between NPCs and tumor stem cells.


Adult Stem Cells/metabolism , Brain/metabolism , Cell Differentiation/physiology , Neural Stem Cells/metabolism , Oncostatin M/metabolism , Adult Stem Cells/cytology , Adult Stem Cells/drug effects , Animals , Brain/cytology , Brain/drug effects , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Cell Separation , Flow Cytometry , Glioblastoma/metabolism , Humans , Immunohistochemistry , Mice , Mice, Inbred C57BL , Mice, Knockout , Neural Stem Cells/cytology , Neural Stem Cells/drug effects , Oncostatin M/pharmacology , Oncostatin M Receptor beta Subunit/metabolism , Polymerase Chain Reaction , Signal Transduction/drug effects , Signal Transduction/physiology
...