Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 32
Filter
Add more filters










Publication year range
1.
Sci Rep ; 14(1): 7742, 2024 04 02.
Article in English | MEDLINE | ID: mdl-38565895

ABSTRACT

Evidence from genetic and epidemiological studies point to lipid metabolism defects in both the brain and periphery being at the core of Alzheimer's disease (AD) pathogenesis. Previously, we reported that central inhibition of the rate-limiting enzyme in monounsaturated fatty acid synthesis, stearoyl-CoA desaturase (SCD), improves brain structure and function in the 3xTg mouse model of AD (3xTg-AD). Here, we tested whether these beneficial central effects involve recovery of peripheral metabolic defects, such as fat accumulation and glucose and insulin handling. As early as 3 months of age, 3xTg-AD mice exhibited peripheral phenotypes including increased body weight and visceral and subcutaneous white adipose tissue as well as diabetic-like peripheral gluco-regulatory abnormalities. We found that intracerebral infusion of an SCD inhibitor that normalizes brain fatty acid desaturation, synapse loss and learning and memory deficits in middle-aged memory-impaired 3xTg-AD mice did not affect these peripheral phenotypes. This suggests that the beneficial effects of central SCD inhibition on cognitive function are not mediated by recovery of peripheral metabolic abnormalities. Given the widespread side-effects of systemically administered SCD inhibitors, these data suggest that selective inhibition of SCD in the brain may represent a clinically safer and more effective strategy for AD.


Subject(s)
Alzheimer Disease , Stearoyl-CoA Desaturase , Mice , Animals , Stearoyl-CoA Desaturase/genetics , Stearoyl-CoA Desaturase/metabolism , Alzheimer Disease/drug therapy , Alzheimer Disease/genetics , Lipid Metabolism/physiology , Lipogenesis , Disease Models, Animal , Mice, Transgenic
3.
Nat Commun ; 13(1): 2061, 2022 04 20.
Article in English | MEDLINE | ID: mdl-35443751

ABSTRACT

The defining features of Alzheimer's disease (AD) include alterations in protein aggregation, immunity, lipid metabolism, synapses, and learning and memory. Of these, lipid abnormalities are the least understood. Here, we investigate the role of Stearoyl-CoA desaturase (SCD), a crucial regulator of fatty acid desaturation, in AD pathogenesis. We show that inhibiting brain SCD activity for 1-month in the 3xTg mouse model of AD alters core AD-related transcriptomic pathways in the hippocampus, and that it concomitantly restores essential components of hippocampal function, including dendritic spines and structure, immediate-early gene expression, and learning and memory itself. Moreover, SCD inhibition dampens activation of microglia, key mediators of spine loss during AD and the main immune cells of the brain. These data reveal that brain fatty acid metabolism links AD genes to downstream immune, synaptic, and functional impairments, identifying SCD as a potential target for AD treatment.


Subject(s)
Alzheimer Disease , Cognitive Dysfunction , Alzheimer Disease/metabolism , Animals , Cognitive Dysfunction/metabolism , Disease Models, Animal , Fatty Acids/metabolism , Hippocampus/metabolism , Mice , Mice, Transgenic , Stearoyl-CoA Desaturase/genetics , Stearoyl-CoA Desaturase/metabolism
4.
Front Neurosci ; 15: 621076, 2021.
Article in English | MEDLINE | ID: mdl-33841077

ABSTRACT

The ventricular-subventricular zone (V-SVZ) is the principal neurogenic niche in the adult mammalian forebrain. Neural stem/progenitor cell (NSPC) activity within the V-SVZ is controlled by numerous of extrinsic factors, whose downstream effects on NSPC proliferation, survival and differentiation are transduced via a limited number of intracellular signaling pathways. Here, we investigated the relationship between age-related changes in NSPC output and activity of signaling pathways downstream of the epidermal growth factor receptor (EGFR), a major regulator of NSPC activity. Biochemical experiments indicated that age-related decline of NSPC activity in vivo is accompanied by selective deficits amongst various EGFR-induced signal pathways within the V-SVZ niche. Pharmacological loss-of-function signaling experiments with cultured NSPCs revealed both overlap and selectivity in the biological functions modulated by the EGFR-induced PI3K/AKT, MEK/ERK and mTOR signaling modules. Specifically, while all three modules promoted EGFR-mediated NSPC proliferation, only mTOR contributed to NSPC survival and only MEK/ERK repressed NSPC differentiation. Using a gain-of-function in vivo genetic approach, we electroporated a constitutively active EGFR construct into a subpopulation of quiescent, EGFR-negative neural stem cells (qNSCs); this ectopic activation of EGFR signaling enabled qNSCs to divide in 3-month-old early adult mice, but not in mice at middle-age or carrying familial Alzheimer disease mutations. Thus, (i) individual EGFR-induced signaling pathways have dissociable effects on NSPC proliferation, survival, and differentiation, (ii) activation of EGFR signaling is sufficient to stimulate qNSC cell cycle entry during early adulthood, and (iii) the proliferative effects of EGFR-induced signaling are dominantly overridden by anti-proliferative signals associated with aging and Alzheimer's disease.

5.
Life Sci Alliance ; 3(7)2020 07.
Article in English | MEDLINE | ID: mdl-32482782

ABSTRACT

The ventricular epithelium of the adult forebrain is a heterogeneous cell population that is a source of both quiescent and activated neural stem cells (qNSCs and aNSCs, respectively). We genetically targeted a subset of ventricle-contacting, glial fibrillary acidic protein (GFAP)-expressing cells, to study their involvement in qNSC/aNSC-mediated adult neurogenesis. Ventricle-contacting GFAP+ cells were lineage-traced beginning in early adulthood using adult brain electroporation and produced small numbers of olfactory bulb neuroblasts until at least 21 mo of age. Notably, electroporated GFAP+ neurogenic precursors were distinct from both qNSCs and aNSCs: they did not give rise to neurosphere-forming aNSCs in vivo or after extended passaging in vitro and they were not recruited during niche regeneration. GFAP+ cells with these properties included a FoxJ1+GFAP+ subset, as they were also present in an inducible FoxJ1 transgenic lineage-tracing model. Transiently overexpressing Mash1 increased the neurogenic output of electroporated GFAP+ cells in vivo, identifying them as a potentially recruitable population. We propose that the qNSC/aNSC lineage of the adult forebrain coexists with a distinct, minimally expanding subset of GFAP+ neurogenic precursors.


Subject(s)
Cerebral Ventricles/metabolism , Epithelium/metabolism , Gene Targeting , Nerve Growth Factors/genetics , Neural Stem Cells/metabolism , Prosencephalon/metabolism , Adult , Adult Stem Cells/metabolism , Animals , Biomarkers , Cell Differentiation/genetics , Fluorescent Antibody Technique , Gene Expression , Glial Fibrillary Acidic Protein/genetics , Glial Fibrillary Acidic Protein/metabolism , Humans , Mice , Mice, Transgenic , Nerve Growth Factors/metabolism , Neural Stem Cells/cytology , Neurogenesis/genetics , Neurons/cytology , Neurons/metabolism , Olfactory Bulb/cytology , Olfactory Bulb/metabolism , Stem Cell Niche/genetics
6.
Theranostics ; 10(14): 6337-6360, 2020.
Article in English | MEDLINE | ID: mdl-32483456

ABSTRACT

Rationale: Monoacylglycerol lipase (Mgll), a hydrolase that breaks down the endocannabinoid 2-arachidonoyl glycerol (2-AG) to produce arachidonic acid (ARA), is a potential target for neurodegenerative diseases, such as Alzheimer's disease (AD). Increasing evidence shows that impairment of adult neurogenesis by perturbed lipid metabolism predisposes patients to AD. However, it remains unknown what causes aberrant expression of Mgll in AD and how Mgll-regulated lipid metabolism impacts adult neurogenesis, thus predisposing to AD during aging. Here, we identify Mgll as an aging-induced factor that impairs adult neurogenesis and spatial memory in AD, and show that metformin, an FDA-approved anti-diabetic drug, can reduce the expression of Mgll to reverse impaired adult neurogenesis, prevent spatial memory decline and reduce ß-amyloid accumulation. Methods: Mgll expression was assessed in both human AD patient post-mortem hippocampal tissues and 3xTg-AD mouse model. In addition, we used both the 3xTg-AD animal model and the CbpS436A genetic knock-in mouse model to identify that elevated Mgll expression is caused by the attenuation of the aPKC-CBP pathway, involving atypical protein kinase C (aPKC)-stimulated Ser436 phosphorylation of histone acetyltransferase CBP through biochemical methods. Furthermore, we performed in vivo adult neurogenesis assay with BrdU/EdU labelling and Morris water maze task in both animal models following pharmacological treatments to show the key role of Mgll in metformin-corrected neurogenesis and spatial memory deficits of AD through reactivating the aPKC-CBP pathway. Finally, we performed in vitro adult neurosphere assays using both animal models to study the role of the aPKC-CBP mediated Mgll repression in determining adult neural stem/progenitor cell (NPC) fate. Results: Here, we demonstrate that aging-dependent induction of Mgll is observed in the 3xTg-AD model and human AD patient post-mortem hippocampal tissues. Importantly, we discover that elevated Mgll expression is caused by the attenuation of the aPKC-CBP pathway. The accumulation of Mgll in the 3xTg-AD mice reduces the genesis of newborn neurons and perturbs spatial memory. However, we find that metformin-stimulated aPKC-CBP pathway decreases Mgll expression to recover these deficits in 3xTg-AD. In addition, we reveal that elevated Mgll levels in cultured adult NPCs from both 3xTg-AD and CbpS436A animal models are responsible for their NPC neuronal differentiation deficits. Conclusion: Our findings set the stage for development of a clinical protocol where Mgll would serve as a biomarker in early stages of AD to identify potential metformin-responsive AD patients to restore their neurogenesis and spatial memory.


Subject(s)
Alzheimer Disease/drug therapy , Alzheimer Disease/enzymology , Metformin/pharmacology , Monoacylglycerol Lipases/metabolism , Neurogenesis/drug effects , Spatial Memory/drug effects , Alzheimer Disease/pathology , Animals , Biomarkers/metabolism , CREB-Binding Protein/metabolism , Disease Models, Animal , Female , Hippocampus/drug effects , Hippocampus/metabolism , Humans , Hypoglycemic Agents/pharmacology , Male , Mice , Mice, Transgenic , Protein Kinase C/metabolism
7.
Cell Rep ; 23(9): 2653-2666, 2018 05 29.
Article in English | MEDLINE | ID: mdl-29847796

ABSTRACT

Late-onset sporadic Alzheimer's disease (AD) is the most prevalent form of dementia, but its origin remains poorly understood. The Bmi1/Ring1 protein complex maintains transcriptional repression of developmental genes through histone H2A mono-ubiquitination, and Bmi1 deficiency in mice results in growth retardation, progeria, and neurodegeneration. Here, we demonstrate that BMI1 is silenced in AD brains, but not in those with early-onset familial AD, frontotemporal dementia, or Lewy body dementia. BMI1 expression was also reduced in cortical neurons from AD patient-derived induced pluripotent stem cells but not in neurons overexpressing mutant APP and PSEN1. BMI1 knockout in human post-mitotic neurons resulted in amyloid beta peptide secretion and deposition, p-Tau accumulation, and neurodegeneration. Mechanistically, BMI1 was required to repress microtubule associated protein tau (MAPT) transcription and prevent GSK3beta and p53 stabilization, which otherwise resulted in neurodegeneration. Restoration of BMI1 activity through genetic or pharmaceutical approaches could represent a therapeutic strategy against AD.


Subject(s)
Alzheimer Disease/pathology , Models, Biological , Polycomb Repressive Complex 1/deficiency , Age of Onset , Alzheimer Disease/genetics , Amyloid/metabolism , Brain/metabolism , Brain/pathology , Dementia/metabolism , Dementia/pathology , Glycogen Synthase Kinase 3 beta/metabolism , Humans , Induced Pluripotent Stem Cells/metabolism , Neurons/metabolism , Phosphorylation , Polycomb Repressive Complex 1/genetics , Polycomb Repressive Complex 1/metabolism , Tumor Suppressor Protein p53/metabolism , tau Proteins/metabolism
8.
Front Mol Neurosci ; 11: 126, 2018.
Article in English | MEDLINE | ID: mdl-29706867

ABSTRACT

Environmental enrichment (EE) is a powerful stimulus of brain plasticity and is among the most accessible treatment options for brain disease. In rodents, EE is modeled using multi-factorial environments that include running, social interactions, and/or complex surroundings. Here, we show that running and running-independent EE differentially affect the hippocampal dentate gyrus (DG), a brain region critical for learning and memory. Outbred male CD1 mice housed individually with a voluntary running disk showed improved spatial memory in the radial arm maze compared to individually- or socially-housed mice with a locked disk. We therefore used RNA sequencing to perform an unbiased interrogation of DG gene expression in mice exposed to either a voluntary running disk (RUN), a locked disk (LD), or a locked disk plus social enrichment and tunnels [i.e., a running-independent complex environment (CE)]. RNA sequencing revealed that RUN and CE mice showed distinct, non-overlapping patterns of transcriptomic changes versus the LD control. Bio-informatics uncovered that the RUN and CE environments modulate separate transcriptional networks, biological processes, cellular compartments and molecular pathways, with RUN preferentially regulating synaptic and growth-related pathways and CE altering extracellular matrix-related functions. Within the RUN group, high-distance runners also showed selective stress pathway alterations that correlated with a drastic decline in overall transcriptional changes, suggesting that excess running causes a stress-induced suppression of running's genetic effects. Our findings reveal stimulus-dependent transcriptional signatures of EE on the DG, and provide a resource for generating unbiased, data-driven hypotheses for novel mediators of EE-induced cognitive changes.

9.
Exp Cell Res ; 368(1): 84-100, 2018 07 01.
Article in English | MEDLINE | ID: mdl-29689278

ABSTRACT

Development of the spinal cord requires dynamic and tightly controlled expression of numerous transcription factors. Forkhead Box protein J1 (FoxJ1) is a transcription factor involved in ciliogenesis and is specifically expressed in ependymal cells (ECs) in the adult central nervous system. However, using FoxJ1 fate-mapping mouse lines, we observed that FoxJ1 is also transiently expressed by the progenitors of other neural subtypes during development. Moreover, using a knock-in mouse line, we discovered that FoxJ1 is essential for embryonic progenitors to follow a normal developmental trajectory. FoxJ1 loss perturbed embryonic progenitor proliferation and cell fate determination, and resulted in formation of adult ECs having impaired stem cell potential and an inability to respond to spinal cord injury in both male and female animals. Thus, our study uncovers unexpected developmental functions of FoxJ1 in cell fate determination of subsets of neural cells and suggests that FoxJ1 is critical for maintaining the stem cell potential of ECs into adulthood.


Subject(s)
Cell Differentiation/physiology , Forkhead Transcription Factors/metabolism , Gene Expression Regulation/genetics , Stem Cells/cytology , Animals , Ependyma/metabolism , Female , Male , Mice , Organogenesis/physiology , Spinal Cord/metabolism , Spinal Cord Injuries/metabolism
10.
Biol Cell ; 110(1): 6-25, 2018 Jan.
Article in English | MEDLINE | ID: mdl-28980327

ABSTRACT

Neural stem cell (NSC) activity and adult neurogenesis are physiologically relevant regulators of adult brain structure, function and repair. Given these roles, the NSC impairments observed in a wide range of neurodegenerative and psychiatric conditions likely factor into the overall cognitive dysfunction in these conditions. We investigated NSC regulation in the context of Alzheimer's disease (AD) using the well-characterised triple transgenic (3xTg) model of AD. In this review, we describe our recent findings that link 3xTg-AD neurogenesis impairments to AD-associated abnormalities in brain fatty acid metabolism. Notably, we identified an accumulation of triglycerides rich in oleic acid, a mono-unsaturated fatty acid, within the forebrain NSC niche in AD. Inhibiting the local conversion of saturated to mono-unsaturated fatty acids within the brain was sufficient to counteract the loss of NSC activity in 3xTg-AD mice (Hamilton et al., 2015). We place these findings within the context of recent evidence that dynamic changes in lipid metabolism occur during the transition from NSC quiescence to activation. The picture that emerges is that the critical NSC quiescence-to-activation decision is sensitive to the local levels of specific fatty acids and can be impaired by a disease-associated shift in brain fatty acid balance.


Subject(s)
Alzheimer Disease/pathology , Brain/pathology , Fatty Acids/metabolism , Neural Stem Cells/pathology , Alzheimer Disease/metabolism , Alzheimer Disease/physiopathology , Animals , Brain/metabolism , Brain/physiopathology , Disease Models, Animal , Fatty Acids/analysis , Humans , Mice, Transgenic , Neural Stem Cells/metabolism , Neurogenesis , Oleic Acid/analysis , Oleic Acid/metabolism
11.
EBioMedicine ; 13: 55-65, 2016 Nov.
Article in English | MEDLINE | ID: mdl-27818039

ABSTRACT

Stem cells have a high therapeutic potential for the treatment of spinal cord injury (SCI). We have shown previously that endogenous stem cell potential is confined to ependymal cells in the adult spinal cord which could be targeted for non-invasive SCI therapy. However, ependymal cells are an understudied cell population. Taking advantage of transgenic lines, we characterize the appearance and potential of ependymal cells during development. We show that spinal cord stem cell potential in vitro is contained within these cells by birth. Moreover, juvenile cultures generate more neurospheres and more oligodendrocytes than adult ones. Interestingly, juvenile ependymal cells in vivo contribute to glial scar formation after severe but not mild SCI, due to a more effective sealing of the lesion by other glial cells. This study highlights the importance of the age-dependent potential of stem cells and post-SCI environment in order to utilize ependymal cell's regenerative potential.


Subject(s)
Cell Differentiation , Ependyma/cytology , Neural Stem Cells/cytology , Regeneration , Spinal Cord Injuries/pathology , Animals , Cell Self Renewal , Cells, Cultured , Disease Models, Animal , Genes, Reporter , Macrophages/immunology , Macrophages/metabolism , Mice , Microglia/immunology , Microglia/metabolism , Neural Stem Cells/metabolism , Spinal Cord/cytology , Spinal Cord/embryology , Spinal Cord/metabolism , Spinal Cord Injuries/immunology , Spinal Cord Injuries/metabolism , Spinal Cord Injuries/therapy
12.
Front Neurosci ; 9: 407, 2015.
Article in English | MEDLINE | ID: mdl-26576147

ABSTRACT

A single asymmetric division by an adult neural stem cell (NSC) ultimately generates dozens of differentiated progeny, a feat made possible by the proliferative expansion of transit-amplifying progenitor cells (TAPs). Although NSC activation and TAP expansion is determined by pro- and anti-proliferative signals found within the niche, remarkably little is known about how these cells integrate simultaneous conflicting signals. We investigated this question focusing on the subventricular zone (SVZ) niche of the adult murine forebrain. Using primary cultures of SVZ cells, we demonstrate that Epidermal Growth Factor (EGF) and Bone Morphogenetic Protein (BMP)-2 are particularly powerful pro- and anti-proliferative factors for SVZ-derived neural precursors. Dose-response experiments showed that when simultaneously exposed to both signals, BMP dominantly suppressed EGF-induced proliferation; moreover, this dominance extended to all parameters of neural precursor behavior tested, including inhibition of proliferation, modulation of cell cycle, promotion of differentiation, and increase of cell death. BMP's anti-proliferative effect did not involve inhibition of mTORC1 or ERK signaling, key mediators of EGF-induced proliferation, and had distinct stage-specific consequences, promoting TAP differentiation but NSC quiescence. In line with these in vitro data, in vivo experiments showed that exogenous BMP limits EGF-induced proliferation of TAPs while inhibition of BMP-SMAD signaling promotes activation of quiescent NSCs. These findings clarify the stage-specific effects of BMPs on SVZ neural precursors, and support a hierarchical model in which the anti-proliferative effects of BMP dominate over EGF proliferation signaling to constitutively drive TAP differentiation and NSC quiescence.

13.
Cell Stem Cell ; 17(4): 397-411, 2015 Oct 01.
Article in English | MEDLINE | ID: mdl-26321199

ABSTRACT

Lipid metabolism is fundamental for brain development and function, but its roles in normal and pathological neural stem cell (NSC) regulation remain largely unexplored. Here, we uncover a fatty acid-mediated mechanism suppressing endogenous NSC activity in Alzheimer's disease (AD). We found that postmortem AD brains and triple-transgenic Alzheimer's disease (3xTg-AD) mice accumulate neutral lipids within ependymal cells, the main support cell of the forebrain NSC niche. Mass spectrometry and microarray analyses identified these lipids as oleic acid-enriched triglycerides that originate from niche-derived rather than peripheral lipid metabolism defects. In wild-type mice, locally increasing oleic acid was sufficient to recapitulate the AD-associated ependymal triglyceride phenotype and inhibit NSC proliferation. Moreover, inhibiting the rate-limiting enzyme of oleic acid synthesis rescued proliferative defects in both adult neurogenic niches of 3xTg-AD mice. These studies support a pathogenic mechanism whereby AD-induced perturbation of niche fatty acid metabolism suppresses the homeostatic and regenerative functions of NSCs.


Subject(s)
Lipid Metabolism , Neural Stem Cells , Prosencephalon/metabolism , Adult Stem Cells/metabolism , Adult Stem Cells/pathology , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Animals , Autopsy , Cell Proliferation , Disease Models, Animal , Mass Spectrometry , Mice , Microarray Analysis , Neural Stem Cells/metabolism , Neural Stem Cells/pathology , Oleic Acid/biosynthesis , Regeneration , Stem Cell Niche
14.
Glia ; 63(8): 1469-82, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25921491

ABSTRACT

Stroke and spinal cord injury (SCI) are among the most frequent causes of central nervous system (CNS) dysfunction, affecting millions of people worldwide each year. The personal and financial costs for affected individuals, their families, and the broader communities are enormous. Although the mammalian CNS exhibits little spontaneous regeneration and self-repair, recent discoveries have revealed that subpopulations of glial cells in the adult forebrain subventricular zone and the spinal cord ependymal zone possess neural stem cell properties. These endogenous neural stem cells react to stroke and SCI by contributing a significant number of new neural cells to formation of the glial scar. These findings have raised hopes that new therapeutic strategies can be designed based on appropriate modulation of endogenous neural stem cell responses to CNS injury. Here, we review the responses of forebrain and spinal cord neural stem cells to stroke and SCI, the role of these responses in restricting injury-induced tissue loss, and the possibility of directing these responses to promote anatomical and functional repair of the CNS.


Subject(s)
Brain Ischemia/physiopathology , Neural Stem Cells/physiology , Spinal Cord Injuries/physiopathology , Stroke/physiopathology , Animals , Brain Ischemia/therapy , Ependyma/physiopathology , Humans , Spinal Cord Injuries/therapy , Stem Cell Niche/physiology , Stroke/therapy
15.
PLoS One ; 9(1): e86237, 2014.
Article in English | MEDLINE | ID: mdl-24465980

ABSTRACT

Environmental enrichment (EE) exerts powerful effects on brain physiology, and is widely used as an experimental and therapeutic tool. Typical EE paradigms are multifactorial, incorporating elements of physical exercise, environmental complexity, social interactions and stress, however the specific contributions of these variables have not been separable using conventional housing paradigms. Here, we evaluated the impacts of these individual variables on adult hippocampal neurogenesis by using a novel "Alternating EE" paradigm. For 4 weeks, adult male CD1 mice were alternated daily between two enriched environments; by comparing groups that differed in one of their two environments, the individual and combinatorial effects of EE variables could be resolved. The Alternating EE paradigm revealed that (1) voluntary running for 3 days/week was sufficient to increase both mitotic and post-mitotic stages of hippocampal neurogenesis, confirming the central importance of exercise; (2) a complex environment (comprised of both social interactions and rotated inanimate objects) had no effect on neurogenesis itself, but enhanced depolarization-induced c-Fos expression (attributable to social interactions) and buffered stress-induced plasma corticosterone levels (attributable to inanimate objects); and (3) neither social isolation, group housing, nor chronically increased levels of plasma corticosterone had a prolonged impact on neurogenesis. Mouse strain, handling and type of running apparatus were tested and excluded as potential confounding factors. These findings provide valuable insights into the relative effects of key EE variables on adult neurogenesis, and this "Alternating EE" paradigm represents a useful tool for exploring the contributions of individual EE variables to mechanisms of neural plasticity.


Subject(s)
Dentate Gyrus/physiopathology , Neurogenesis , Running , Stress, Psychological/physiopathology , Animals , Behavior, Animal , Corticosterone/blood , Dentate Gyrus/metabolism , Dentate Gyrus/pathology , Environment , Gene Expression , Housing, Animal , Male , Membrane Potentials , Mice , Mice, Inbred C57BL , Proto-Oncogene Proteins c-fos/genetics , Proto-Oncogene Proteins c-fos/metabolism , Social Behavior , Stress, Psychological/metabolism , Stress, Psychological/pathology
16.
PLoS One ; 9(1): e85916, 2014.
Article in English | MEDLINE | ID: mdl-24475059

ABSTRACT

The adult mammalian spinal cord has limited regenerative capacity in settings such as spinal cord injury (SCI) and multiple sclerosis (MS). Recent studies have revealed that ependymal cells lining the central canal possess latent neural stem cell potential, undergoing proliferation and multi-lineage differentiation following experimental SCI. To determine whether reactive ependymal cells are a realistic endogenous cell population to target in order to promote spinal cord repair, we assessed the spatiotemporal dynamics of ependymal cell proliferation for up to 35 days in three models of spinal pathologies: contusion SCI using the Infinite Horizon impactor, focal demyelination by intraspinal injection of lysophosphatidylcholine (LPC), and autoimmune-mediated multi-focal demyelination using the active experimental autoimmune encephalomyelitis (EAE) model of MS. Contusion SCI at the T9-10 thoracic level stimulated a robust, long-lasting and long-distance wave of ependymal proliferation that peaked at 3 days in the lesion segment, 14 days in the rostral segment, and was still detectable at the cervical level, where it peaked at 21 days. This proliferative wave was suppressed distal to the contusion. Unlike SCI, neither chemical- nor autoimmune-mediated demyelination triggered ependymal cell proliferation at any time point, despite the occurrence of demyelination (LPC and EAE), remyelination (LPC) and significant locomotor defects (EAE). Thus, traumatic SCI induces widespread and enduring activation of reactive ependymal cells, identifying them as a robust cell population to target for therapeutic manipulation after contusion; conversely, neither demyelination, remyelination nor autoimmunity appears sufficient to trigger proliferation of quiescent ependymal cells in models of MS-like demyelinating diseases.


Subject(s)
Cell Proliferation , Ependyma/cytology , Neural Stem Cells/cytology , Spinal Canal/cytology , Spinal Cord Injuries/physiopathology , Adult , Analysis of Variance , Animals , Encephalomyelitis, Autoimmune, Experimental , Female , Humans , Immunohistochemistry , Laminectomy , Lysophosphatidylcholines , Mice , Mice, Inbred C57BL , Microscopy, Fluorescence , Neural Stem Cells/physiology
17.
Eur J Neurosci ; 37(12): 1978-86, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23773067

ABSTRACT

In the brains of adult vertebrates, including humans, neurogenesis occurs in restricted niches where it maintains cellular turnover and cognitive plasticity. In virtually all species, however, aging is associated with a significant decline in adult neurogenesis. Moreover, an acceleration of neurogenic defects is observed in models of Alzheimer's disease and other neurodegenerative diseases, suggesting an involvement in aging- and disease-associated cognitive deficits. To gain insights into when, how and why adult neurogenesis decreases in the aging brain, we critically reviewed the scientific literature on aging of the rodent subventricular zone, the neurogenic niche of the adult forebrain. Our analysis revealed that deficits in the neurogenic pathway are largely established by middle age, but that there remains striking ambiguity in the underlying mechanisms, especially at the level of stem and progenitor cells. We identify and discuss several challenging issues that have contributed to these key gaps in our current knowledge. In the future, addressing these issues should help untangle the interactions between neurogenesis, aging and aging-associated diseases.


Subject(s)
Aging/physiology , Lateral Ventricles/cytology , Neural Stem Cells/cytology , Neurogenesis/physiology , Prosencephalon/cytology , Alzheimer Disease/pathology , Animals , Mice , Rats
18.
J Cell Physiol ; 228(9): 1844-53, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23459851

ABSTRACT

The rodent heart contains a population of nestin((+)) cells derived from the embryonic neural crest and migrate to the scar after myocardial infarction (MI). The present study tested the hypothesis that intron 2 of the nestin gene drives expression and a subpopulation of nestin((+)) cells participate in reparative vascularisation. The directed expression of the green fluorescent protein (GFP) by the second intron of the nestin gene identified GFP/nestin((+)) cells intercalated among ventricular myocytes in the heart of normal transgenic mice. Ischemic injury led to the migration of GFP((+)) cells to the scar and a subpopulation was detected in CD31/nestin((+)) endothelial cells of newly formed blood vessels. The direct contribution to reparative vascularisation provided the impetus to test the hypothesis that increasing the population of nestin((+)) cells in the infarcted heart will improve scar healing. Skin-derived cells isolated from E18 Sprague-Dawley rats grew as spheres, expressed nestin, sox2, neural crest-related transcriptional genes and a panel of peptide growth factors. Skin-derived cells transplanted in the non-infarcted left ventricle of 3-day post-MI rats migrated to the peri-infarct/infarct region and remained engrafted for 21 days. A significantly smaller infarct, increased number of small calibre blood vessels and improved ventricular function were observed in engrafted infarcted rat hearts. Thus, the second intron of the nestin gene drives expression in the mouse heart and a subpopulation of GFP/nestin((+)) cells directly participate in reparative vascularisation. Increasing the population of nestin((+)) cells via the transplantation of skin-derived cells represents a potential approach to limit ischemic damage to the heart.


Subject(s)
Intermediate Filament Proteins/genetics , Myocytes, Cardiac/metabolism , Neovascularization, Physiologic/genetics , Nerve Tissue Proteins/genetics , Neural Crest/growth & development , Animals , Cell Differentiation , Intermediate Filament Proteins/metabolism , Male , Mice , Mice, Transgenic , Myocardial Infarction/metabolism , Myocardial Infarction/physiopathology , Myocardium/cytology , Myocardium/metabolism , Myocytes, Cardiac/cytology , Myocytes, Cardiac/physiology , Nerve Tissue Proteins/metabolism , Nestin , Neural Crest/cytology , Neural Crest/metabolism , Neurons/cytology , Neurons/metabolism , Rats , Rats, Sprague-Dawley , Stem Cells/cytology , Stem Cells/metabolism
19.
J Neurosci ; 32(43): 15012-26, 2012 Oct 24.
Article in English | MEDLINE | ID: mdl-23100423

ABSTRACT

Adult forebrain neurogenesis is dynamically regulated. Multiple families of niche-derived cues have been implicated in this regulation, but the precise roles of key intracellular signaling pathways remain vaguely defined. Here, we show that mammalian target of rapamycin (mTOR) signaling is pivotal in determining proliferation versus quiescence in the adult forebrain neural stem cell (NSC) niche. Within this niche, mTOR complex-1 (mTORC1) activation displays stage specificity, occurring in transiently amplifying (TA) progenitor cells but not in GFAP+ stem cells. Inhibiting mTORC1 depletes the TA progenitor pool in vivo and suppresses epidermal growth factor (EGF)-induced proliferation within neurosphere cultures. Interestingly, mTORC1 inhibition induces a quiescence-like phenotype that is reversible. Likewise, mTORC1 activity and progenitor proliferation decline within the quiescent NSC niche of the aging brain, while EGF administration reactivates the quiescent niche in an mTORC1-dependent manner. These findings establish fundamental links between mTOR signaling, proliferation, and aging-associated quiescence in the adult forebrain NSC niche.


Subject(s)
Aging , Cell Differentiation/physiology , Neural Stem Cells/physiology , Prosencephalon/cytology , Signal Transduction/physiology , TOR Serine-Threonine Kinases/metabolism , 2',3'-Cyclic-Nucleotide Phosphodiesterases/metabolism , Animals , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Differentiation/drug effects , Cell Differentiation/genetics , Cells, Cultured , Doublecortin Domain Proteins , Embryo, Mammalian , Female , Fibroblast Growth Factor 2/pharmacology , Fibroblast Growth Factors/pharmacology , Gene Expression Regulation, Developmental/genetics , Glial Fibrillary Acidic Protein/genetics , Green Fluorescent Proteins/genetics , Humans , Ki-67 Antigen/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microdissection , Microtubule-Associated Proteins/metabolism , Nerve Growth Factors/metabolism , Nerve Tissue Proteins/metabolism , Neural Stem Cells/drug effects , Neuropeptides/metabolism , Oligodendrocyte Transcription Factor 2 , Pregnancy , Ribosomal Protein S6/metabolism , S100 Calcium Binding Protein beta Subunit , S100 Proteins/metabolism , Signal Transduction/drug effects , Signal Transduction/genetics , Sirolimus/metabolism , TOR Serine-Threonine Kinases/genetics , Transfection , Tubulin/metabolism
20.
Genes Chromosomes Cancer ; 51(8): 792-804, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22514116

ABSTRACT

Neuroblastoma (NB) is the most common and lethal extracranial solid tumor of childhood. Despite aggressive therapy, more than half of the children with advanced NB will die of uncontrolled metastatic disease. After chemotherapy, tumor-initiating cells (TICs) could persist, cause relapses and metastasis. The aim of this study is to demonstrate the tumor-initiating properties of CD133high NB cells and to identify new specific genetic abnormalities. Isolation of the CD133high cell population from NB cell lines was followed by neurosphere formation, soft agar assays, and orthotopic injections in NOD/SCID/IL2Rγc-null mice. A differential genotyping analysis was performed with Affymetrix SNP 6.0 arrays on CD133low and CD133high populations and the frequency of the abnormalities of 36 NB tumors was determined. Our results show that CD133high NB cells possess tumor-initiating properties, as CD133high cells formed significantly more neurospheres and produced significantly more colonies in soft agar than CD133low. Injection of 500 CD133high cells was sufficient to generate primary tumors and frequent metastases in mice. Differential genotyping analysis demonstrated two common regions with gains (16p13.3 and 19p13.3) including the gene EFNA2 in the CD133high population, and two with loss of heterozygosity (16q12.1 and 21q21.3) in the CD133low population. The gain of EFNA2 correlated with increased expression of the corresponding protein. These abnormalities were found in NB samples and some were significantly correlated with CD133 expression. Our results show that CD133high NB cells have TICs properties and present different genotyping characteristics compared to CD133low cells. Our findings reveal insights into new therapeutic targets in NB TICs.


Subject(s)
Antigens, CD/genetics , Glycoproteins/genetics , Neuroblastoma/genetics , Peptides/genetics , AC133 Antigen , Adrenal Gland Neoplasms , Animals , Antigens, CD/biosynthesis , Antigens, CD/metabolism , Cell Line, Tumor , Cell Separation , Chi-Square Distribution , Chromosome Aberrations , Female , Genotype , Glycoproteins/biosynthesis , Glycoproteins/metabolism , Humans , Male , Mice , Mice, Inbred NOD , Mice, SCID , Mutagenesis, Insertional , Neoplasm Transplantation , Neuroblastoma/metabolism , Peptides/metabolism , Polymorphism, Single Nucleotide
SELECTION OF CITATIONS
SEARCH DETAIL
...