Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 68
1.
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
2.
Neurobiol Aging ; 132: 120-130, 2023 Dec.
Article En | MEDLINE | ID: mdl-37801885

Dysfunction of the cholinergic basal forebrain (BF) system and amyloid-ß (Aß) deposition are early pathological features in Alzheimer's disease (AD). However, their association in early AD is not well-established. This study investigated the nature and magnitude of volume loss in the BF, over an extended period, in 516 older adults who completed Aß-PET and serial magnetic resonance imaging scans. Individuals were grouped at baseline according to the presence of cognitive impairment (CU, CI) and Aß status (Aß-, Aß+). Longitudinal volumetric changes in the BF and hippocampus were assessed across groups. The results indicated that high Aß levels correlated with faster volume loss in the BF and hippocampus, and the effect of Aß varied within BF subregions. Compared to CU Aß+ individuals, Aß-related loss among CI Aß+ adults was much greater in the predominantly cholinergic subregion of Ch4p, whereas no difference was observed for the Ch1/Ch2 region. The findings support early and substantial vulnerability of the BF and further reveal distinctive degeneration of BF subregions during early AD.


Alzheimer Disease , Basal Forebrain , Cognitive Dysfunction , Humans , Aged , Alzheimer Disease/diagnostic imaging , Alzheimer Disease/pathology , Basal Forebrain/diagnostic imaging , Basal Forebrain/pathology , Aging/pathology , Amyloid beta-Peptides , Magnetic Resonance Imaging , Cholinergic Agents , Cognitive Dysfunction/diagnostic imaging , Cognitive Dysfunction/pathology , Positron-Emission Tomography
3.
J Neurochem ; 165(6): 791-808, 2023 06.
Article En | MEDLINE | ID: mdl-36660878

The traditional medicinal mushroom Hericium erinaceus is known for enhancing peripheral nerve regeneration through targeting nerve growth factor (NGF) neurotrophic activity. Here, we purified and identified biologically new active compounds from H. erinaceus, based on their ability to promote neurite outgrowth in hippocampal neurons. N-de phenylethyl isohericerin (NDPIH), an isoindoline compound from this mushroom, together with its hydrophobic derivative hericene A, were highly potent in promoting extensive axon outgrowth and neurite branching in cultured hippocampal neurons even in the absence of serum, demonstrating potent neurotrophic activity. Pharmacological inhibition of tropomyosin receptor kinase B (TrkB) by ANA-12 only partly prevented the NDPIH-induced neurotrophic activity, suggesting a potential link with BDNF signaling. However, we found that NDPIH activated ERK1/2 signaling in the absence of TrkB in HEK-293T cells, an effect that was not sensitive to ANA-12 in the presence of TrkB. Our results demonstrate that NDPIH acts via a complementary neurotrophic pathway independent of TrkB with converging downstream ERK1/2 activation. Mice fed with H. erinaceus crude extract and hericene A also exhibited increased neurotrophin expression and downstream signaling, resulting in significantly enhanced hippocampal memory. Hericene A therefore acts through a novel pan-neurotrophic signaling pathway, leading to improved cognitive performance.


MAP Kinase Signaling System , Spatial Memory , Mice , Animals , Signal Transduction , Neurons/metabolism , Hippocampus/metabolism , Brain-Derived Neurotrophic Factor/pharmacology , Brain-Derived Neurotrophic Factor/metabolism , Receptor, trkB/metabolism , Cells, Cultured
4.
Nat Commun ; 13(1): 6543, 2022 11 02.
Article En | MEDLINE | ID: mdl-36323689

Although epidemiological studies indicate that sleep-disordered breathing (SDB) such as obstructive sleep apnea is a strong risk factor for the development of Alzheimer's disease (AD), the mechanisms of the risk remain unclear. Here we developed a method of modeling SDB in mice that replicates key features of the human condition: altered breathing during sleep, sleep disruption, moderate hypoxemia, and cognitive impairment. When we induced SDB in a familial AD model, the mice displayed exacerbation of cognitive impairment and the pathological features of AD, including increased levels of amyloid-beta and inflammatory markers, as well as selective degeneration of cholinergic basal forebrain neurons. These pathological features were not induced by chronic hypoxia or sleep disruption alone. Our results also revealed that the cholinergic neurodegeneration was mediated by the accumulation of nuclear hypoxia inducible factor 1 alpha. Furthermore, restoring blood oxygen levels during sleep to prevent hypoxia prevented the pathological changes induced by the SDB. These findings suggest a signaling mechanism whereby SDB induces cholinergic basal forebrain degeneration.


Alzheimer Disease , Basal Forebrain , Sleep Apnea Syndromes , Animals , Mice , Humans , Alzheimer Disease/pathology , Basal Forebrain/pathology , Disease Models, Animal , Sleep Apnea Syndromes/complications , Hypoxia/pathology , Cholinergic Agents
5.
Neurobiol Aging ; 117: 24-32, 2022 09.
Article En | MEDLINE | ID: mdl-35640461

Degeneration of cholinergic neurons in the basal forebrain (BF) contributes to cognitive impairment in Alzheimer's disease (AD) and other disorders. Atrophy of BF volume measured by structural MRI is thought to represent the loss of cholinergic neurons in this structure. As there are multiple types of neurons in the BF as well as glia and axons, whether this MRI measure actually reflects the change of cholinergic neurons has not been verified. In this study, we assessed BF cholinergic neuron number by histological counts and compared with the volume measurements by in vivo MRI in 3xTg mice, a model of familial AD. Both manual and template-based segmentation revealed atrophy of the medial septum (MS), consistent with a significant reduction in cholinergic neuron number. However, MRI-measured volume reduction did not correlate with the reduced cholinergic neuron number. To directly test whether specific loss of cholinergic neurons results in BF atrophy, we selectively ablated the cholinergic neurons in the MS. However, no detectable change in MRI volume was observed between lesioned and unlesioned mice. The results indicate that although loss of cholinergic neurons within the BF likely contributes to volume loss, this volume change cannot be taken as a direct biomarker of cholinergic neuron number.


Alzheimer Disease , Basal Forebrain , Alzheimer Disease/diagnostic imaging , Alzheimer Disease/pathology , Animals , Atrophy/pathology , Basal Forebrain/diagnostic imaging , Basal Forebrain/pathology , Cholinergic Agents , Cholinergic Neurons/pathology , Disease Models, Animal , Magnetic Resonance Imaging/methods , Mice
6.
Neuroimage Clin ; 34: 102992, 2022.
Article En | MEDLINE | ID: mdl-35344804

Dysfunction of the cholinergic basal forebrain (BF) neurotransmitter system, including cholinergic axon denervation of the cortex, plays an important role in cognitive decline and dementia. A validated method to directly quantify cortical cholinergic terminal integrity enables exploration of the involvement of this system in diverse cognitive profiles associated with dementia, particularly at a prodromal stage. In this study, we used the radiotracer [18F]-fluoroethoxybenzovesamicol (FEOBV) as a direct measure of cholinergic terminal integrity and investigated its value for the assessment of cholinergic denervation in the cortex and associated cognitive deficits. Eighteen participants (8 with mild cognitive impairment (MCI) and 10 cognitively unimpaired controls) underwent neuropsychological assessment and brain imaging using FEOBV and [18F]-florbetaben for amyloid-ß imaging. The MCI group showed a significant global reduction of FEOBV retention in the cortex and in the parietal and occipital cortices specifically compared to the control group. The global cortical FEOBV retention of all participants positively correlated with the BF, hippocampus and grey matter volumes, but no association was found between the global FEOBV retention and amyloid-ß status. Topographic profiles from voxel-wise analysis of FEOBV images revealed significant positive correlations with the cognitive domains associated with the underlying cortical areas. Overlapping profiles of decreased FEOBV were identified in correlation with impairment in executive function, attention and language, which covered the anterior cingulate gyrus, olfactory cortex, calcarine cortex, middle temporal gyrus and caudate nucleus. However, the absence of cortical atrophy in these areas suggested that reduced cholinergic terminal integrity in the cortex is an important factor underlying the observed cognitive decline in early dementia. Our results provide support for the utility and validity of FEOBV PET for quantitative assessment of region-specific cholinergic terminal integrity that could potentially be used for early detection of cholinergic dysfunction in dementia following further validation in larger cohorts.


Alzheimer Disease , Basal Forebrain , Cognitive Dysfunction , Dementia , Alzheimer Disease/diagnosis , Amyloid beta-Peptides , Cholinergic Agents , Cognitive Dysfunction/diagnostic imaging , Humans , Magnetic Resonance Imaging , Piperidines , Positron-Emission Tomography/methods
7.
J Biol Chem ; 298(3): 101568, 2022 03.
Article En | MEDLINE | ID: mdl-35051416

Neurotrophin signaling is essential for normal nervous system development and adult function. Neurotrophins are secreted proteins that signal via interacting with two neurotrophin receptor types: the multifaceted p75 neurotrophin receptor and the tropomyosin receptor kinase receptors. In vivo, neurons compete for the limited quantities of neurotrophins, a process that underpins neural plasticity, axonal targeting, and ultimately survival of the neuron. Thirty years ago, it was discovered that p75 neurotrophin receptor and tropomyosin receptor kinase A form a complex and mediate high-affinity ligand binding and survival signaling; however, despite decades of functional and structural research, the mechanism of modulation that yields this high-affinity complex remains unclear. Understanding the structure and mechanism of high-affinity receptor generation will allow development of pharmaceuticals to modulate this function for treatment of the many nervous system disorders in which altered neurotrophin expression or signaling plays a causative or contributory role. Here we re-examine the key older literature and integrate it with more recent studies on the topic of how these two receptors interact. We also identify key outstanding questions and propose a model of inside-out allosteric modulation to assist in resolving the elusive high-affinity mechanism and complex.


Receptor, Nerve Growth Factor , Receptor, trkA , Tropomyosin , Animals , Humans , Nerve Growth Factors/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , Receptor, Nerve Growth Factor/metabolism , Receptor, trkA/genetics , Receptor, trkA/metabolism , Receptors, Nerve Growth Factor
8.
Sci Adv ; 8(3): eabj2797, 2022 01 21.
Article En | MEDLINE | ID: mdl-35044824

Inappropriate expansion of antibody-secreting cells (ASCs) is typical of systemic lupus erythematosus (SLE), but the regulatory signaling of pathogenic ASCs is unclear. The present study shows that brain-derived neurotrophic factor precursor (proBDNF) and its high-affinity pan-75 neurotrophin receptor (p75NTR) are highly expressed in CD19+CD27hiCD38hi ASCs in patients with SLE and in CD19+CD44hiCD138+ ASCs in lupus-like mice. The increased proBDNF+ ASCs were positively correlated with clinical symptoms and higher titers of autoantibodies in SLE. Administration of monoclonal antibodies against proBDNF or specific knockout of p75NTR in CD19+ B cells exerted a therapeutic effect on lupus mice by limiting the proportion of ASCs, reducing the production of autoantibodies and attenuating kidney injury. Blocking the biological function of proBDNF or p75NTR also inhibits ASC differentiation and antibody production in vitro. Together, these findings suggest that proBDNF-p75NTR signaling plays a critical pathogenic role in SLE through promoting ASC dysfunction.


Lupus Erythematosus, Systemic , Receptors, Nerve Growth Factor , Animals , Antigens, CD19 , Autoantibodies , B-Lymphocytes , Brain-Derived Neurotrophic Factor/metabolism , Humans , Mice , Mice, Inbred C57BL , Receptors, Nerve Growth Factor/genetics , Receptors, Nerve Growth Factor/metabolism , Up-Regulation
9.
J Psychiatr Res ; 147: 166-175, 2022 03.
Article En | MEDLINE | ID: mdl-35051715

OBJECTIVE: Emerging evidence suggests that people with severe mental illness (SMI) have an increased risk of frailty. We conducted a systematic review to investigate the prevalence and correlates of frailty, as well as the efficacy of frailty interventions, in this population. METHODS: We searched databases from inception to 21 September 2021 for studies that assessed or intervened for frailty in relation to an SMI diagnosis. A narrative synthesis explored the characteristics and adverse health outcomes associated with frailty and the efficacy of interventions. The prevalence of frailty was investigated, and its relationship with age was analysed by a meta-regression. RESULTS: Twenty-five studies involving 2499 patients, primarily older adults, were included in the narrative synthesis. Frailty was associated with higher rates of physical comorbidity, cognitive deficits, falls and mortality among those with SMI. The efficacy of a yoga intervention was investigated in one study, without sustained reductions in frailty. The prevalence of frailty varied between 10.2 and 89.7% and was high in comparison to the general population. CONCLUSIONS: The prevalence of frailty was high in those with SMI and ranged widely due to heterogeneity of study populations. Assessing frailty enables the identification of patients who could benefit from interventions and assists in treatment-related decision making. Further research is required to develop appropriate frailty interventions for this population.


Frailty , Mental Disorders , Aged , Comorbidity , Frailty/epidemiology , Humans , Mental Disorders/epidemiology , Mental Disorders/therapy , Prevalence
10.
Mol Neurodegener ; 17(1): 2, 2022 01 09.
Article En | MEDLINE | ID: mdl-35000606

The use of animals as models of human physiology is, and has been for many years, an indispensable tool for understanding the mechanisms of human disease. In Parkinson's disease, various mouse models form the cornerstone of these investigations. Early models were developed to reflect the traditional histological features and motor symptoms of Parkinson's disease. However, it is important that models accurately encompass important facets of the disease to allow for comprehensive mechanistic understanding and translational significance. Circadian rhythm and sleep issues are tightly correlated to Parkinson's disease, and often arise prior to the presentation of typical motor deficits. It is essential that models used to understand Parkinson's disease reflect these dysfunctions in circadian rhythms and sleep, both to facilitate investigations into mechanistic interplay between sleep and disease, and to assist in the development of circadian rhythm-facing therapeutic treatments. This review describes the extent to which various genetically- and neurotoxically-induced murine models of Parkinson's reflect the sleep and circadian abnormalities of Parkinson's disease observed in the clinic.


Parkinson Disease , REM Sleep Behavior Disorder , Animals , Circadian Rhythm , Mice , Parkinson Disease/diagnosis , REM Sleep Behavior Disorder/diagnosis , Sleep
11.
J Neurochem ; 158(6): 1292-1306, 2021 09.
Article En | MEDLINE | ID: mdl-34109634

Cholinergic basal forebrain (cBF) neurons are particularly vulnerable to degeneration following trauma and in neurodegenerative conditions. One reason for this is their characteristic expression of the p75 neurotrophin receptor (p75NTR ), which is up-regulated and mediates neuronal death in a range of neurological and neurodegenerative conditions, including dementia, stroke and ischaemia. The signalling pathway by which p75NTR signals cell death is incompletely characterised, but typically involves activation by neurotrophic ligands and signalling through c-Jun kinase, resulting in caspase activation via mitochondrial apoptotic signalling pathways. Less well appreciated is the link between conditions of oxidative stress and p75NTR death signalling. Here, we review the literature describing what is currently known regarding p75NTR death signalling in environments of oxidative stress and hypoxia to highlight the overlap in signalling pathways and the implications for p75NTR signalling in cBF neurons. We propose that there is a causal relationship and define key questions to test this assertion.


Cholinergic Neurons/metabolism , Hypoxia/metabolism , Nerve Degeneration/metabolism , Nerve Tissue Proteins/metabolism , Oxidative Stress/physiology , Receptors, Nerve Growth Factor/metabolism , Animals , Basal Forebrain/metabolism , Basal Forebrain/pathology , Cell Death/physiology , Cholinergic Neurons/pathology , Humans , Hypoxia/pathology , Nerve Degeneration/pathology
12.
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
13.
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
14.
iScience ; 24(2): 102043, 2021 Feb 19.
Article En | MEDLINE | ID: mdl-33537655

Major depression is a prevalent and long-lasting psychiatric illness with severe functional impairment and high suicide rate. We have previously shown that the ventrolateral orbital cortex (VLO) plays a key role in the stress responses in mice, but the underlying mechanisms remains unclear. Here, we used proteomic method to identify differentially expressed proteins in VLO of chronic unpredictable mild stress (CUMS) mice. Of 4,953 quantified proteins, 45 proteins were differentially expressed following CUMS. The integrated pathway analyses identified 14-3-3ε and TrkB signaling as differentially downregulated in association with stress-induced depressive-like behaviors. 14-3-3ε overexpression in VLO relieved the depressive-like behaviors by rescue of Bad-mediated apoptosis. Moreover, treatment with the 14-3-3ε stabilizer FC-A precluded neuronal apoptotic signaling in VLO of depressed mice. Because 14-3-3ε provides significant protection against chronic stress, boosting 14-3-3ε expression, pharmacological stabilization of 14-3-3s (e.g. with FC-A) is identified as an exciting therapeutic target for major depression.

15.
J Neurochem ; 158(3): 807-817, 2021 08.
Article En | MEDLINE | ID: mdl-32628780

The blood-brain barrier (BBB) and blood-spinal cord barrier (BSCB) are highly specialized structures that limit molecule entry from the blood and maintain homeostasis within the central nervous system (CNS). BBB and BSCB breakdown are associated with multiple neurodegenerative diseases. Given the key role of neuroprotective barrier impairment in neurodegeneration, it is important to identify an effective quantitative method to assess barrier integrity in animal models. In this study, we developed and validated a quantitative method for assessing BBB and BSCB integrity using sodium fluorescein, a compound that outperformed other fluorescent dyes. We demonstrated using this method that multiple CNS regions progressively increase in permeability in models of Huntington's disease and amyotrophic lateral sclerosis, whereas biphasic disruption occurred in a mouse model of Alzheimer's disease with disease progression. Collectively, we report a quantitative fluorometric marker with validated reproducible experimental methods that allows the effective assessment of BBB and BSCB integrity in animal models. This method could be useful to further the understanding of the contribution of these neuroprotective barriers to neurodegeneration processes.


Blood-Brain Barrier/metabolism , Brain/metabolism , Fluorometry/standards , Neurodegenerative Diseases/metabolism , Neuroprotection/physiology , Spinal Cord/metabolism , Animals , Blood-Brain Barrier/pathology , Brain/pathology , Disease Models, Animal , Fluorometry/methods , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neurodegenerative Diseases/genetics , Reproducibility of Results , Spinal Cord/pathology
16.
J Neurosci Res ; 98(10): 1987-1998, 2020 10.
Article En | MEDLINE | ID: mdl-32585763

The p75 neurotrophin receptor (p75NTR ) is required for maintaining peripheral sensory neuron survival and function; however, the underlying cellular mechanism remains unclear. The general view is that expression of p75NTR by the neuron itself is required for maintaining sensory neuron survival and myelination in the peripheral nervous system (PNS). Adopting a neuronal-specific conditional knockout strategy, we demonstrate the partial depletion of p75NTR in neurons exerts little influence upon maintaining sensory neuron survival and peripheral nerve myelination in health and after demyelinating neuropathy. Our data show that the density and total number of dorsal root ganglion (DRG) neurons in 2-month-old mice is not affected following the deletion of p75NTR in large-diameter myelinating neurons, as assessed by stereology. Adopting experimental autoimmune neuritis induced in adult male mice, an animal model of demyelinating peripheral neuropathy, we identify that deleting p75NTR in myelinating neurons exerts no influence upon the disease progression, the total number of DRG neurons, and the extent of myelin damage in the sciatic nerve, indicating that the expression of neuronal p75NTR is not essential for maintaining peripheral neuron survival and myelination after a demyelinating insult in vivo. Together, results of this study suggest that the survival and myelination of peripheral sensory neurons is independent of p75NTR expressed by a subtype of neurons in vivo. Thus, our findings provide new insights into the mechanism underpinning p75NTR -mediated neuronal survival in the PNS.


Ganglia, Spinal/metabolism , Receptors, Nerve Growth Factor/deficiency , Receptors, Nerve Growth Factor/genetics , Sensory Receptor Cells/metabolism , Animals , Cell Survival/physiology , Female , Gene Deletion , Male , Mice , Mice, Knockout , Mice, Transgenic
17.
Clin Exp Pharmacol Physiol ; 47(10): 1740-1750, 2020 10.
Article En | MEDLINE | ID: mdl-32542833

In Alzheimer's disease (AD) glial fibrillary acidic protein (GFAP) is expressed by reactive astrocytes surrounding ß-amyloid (Aß) plaques, whereas brain-derived neurotrophic factor (BDNF) levels are typically reduced. We compared the expression of GFAP, BDNF, and its precursor proBDNF in the dorsal hippocampus of two transgenic AD mouse models. APPSwe YAC mice expressing the APPSwe transgene on a yeast artificial chromosome (YAC) were assessed at age 4 and 21 months, and APPSwe/PS1dE9 mice co-expressing mutant amyloid precursor protein (APPSwe) and presenilin-1 (PS1dE9) were assessed at age 4 and 9 months. Significantly increased (1.4-fold) GFAP expression was observed in APPSwe YAC c.f. wild-type (Wt) mice aged 21 months, when Aß deposition was first evident in these mice. In APPSwe/PS1dE9 mice aged 4 and 9 months, GFAP expression was significantly increased (1.6- and 3.1-fold, respectively) c.f. Wt mice, and was associated with robust Aß deposition at 9 months. BDNF expression was significantly lower in 4- and 21-month old APPSwe YAC mice (0.8- and 0.6-fold, respectively) c.f. age-matched Wt mice, whereas proBDNF expression was significantly higher (10-fold) in the APPSwe YAC c.f. Wt mice aged 21 months. In APPSwe/PS1dE9 mice aged 4 months, BDNF expression was significantly lower (0.4-fold) c.f. age-matched Wt mice and was equivalent to that in 9-month old mice of both genotypes; proBDNF expression mirrored that of BDNF in this strain. These findings support a role for reactive astrocytes and neuroinflammation, rather than BDNF, in the spatial memory deficits previously reported for APPSwe YAC and APPSwe/PS1dE9 mice.


Alzheimer Disease/metabolism , Brain-Derived Neurotrophic Factor/metabolism , Brain/metabolism , Glial Fibrillary Acidic Protein/metabolism , Alzheimer Disease/physiopathology , Amyloid beta-Peptides/metabolism , Animals , Brain/physiopathology , Disease Models, Animal , Humans , Maze Learning , Mice , Mice, Transgenic , Spatial Memory
18.
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
19.
Development ; 146(18)2019 09 18.
Article En | MEDLINE | ID: mdl-31488566

During development, the p75 neurotrophin receptor (p75NTR) is widely expressed in the nervous system where it regulates neuronal differentiation, migration and axonal outgrowth. p75NTR also mediates the survival and death of newly born neurons, with functional outcomes being dependent on both timing and cellular context. Here, we show that knockout of p75NTR from embryonic day 10 (E10) in neural progenitors using a conditional Nestin-Cre p75NTR floxed mouse causes increased apoptosis of progenitor cells. By E14.5, the number of Tbr2-positive progenitor cells was significantly reduced and the rate of neurogenesis was halved. Furthermore, in adult knockout mice, there were fewer cortical pyramidal neurons, interneurons, cholinergic basal forebrain neurons and striatal neurons, corresponding to a relative reduction in volume of these structures. Thalamic midline fusion during early postnatal development was also impaired in Nestin-Cre p75NTR floxed mice, indicating a novel role for p75NTR in the formation of this structure. The phenotype of this strain demonstrates that p75NTR regulates multiple aspects of brain development, including cortical progenitor cell survival, and that expression during early neurogenesis is required for appropriate formation of telencephalic structures.


Basal Forebrain/embryology , Neocortex/embryology , Neostriatum/embryology , Neural Stem Cells/cytology , Neural Stem Cells/metabolism , Receptor, Nerve Growth Factor/metabolism , Thalamus/embryology , Animals , Animals, Newborn , Caspase 3/metabolism , Cell Proliferation , Cell Survival , Golgi Apparatus/metabolism , Interneurons/metabolism , Mice , Nestin/metabolism , Neurogenesis , Neurons/cytology , Neurons/metabolism , Organ Size , Pyramidal Cells/metabolism
20.
J Neurosci ; 39(40): 7976-7991, 2019 10 02.
Article En | MEDLINE | ID: mdl-31363064

Alzheimer's disease (AD) is associated with the cleavage of the amyloid precursor protein (APP) to produce the toxic amyloid-ß (Aß) peptide. Accumulation of Aß, together with the concomitant inflammatory response, ultimately leads to neuronal death and cognitive decline. Despite AD progression being underpinned by both neuronal and immunological components, therapeutic strategies based on dual targeting of these systems remains unexplored. Here, we report that inactivation of the p110δ isoform of phosphoinositide 3-kinase (PI3K) reduces anterograde axonal trafficking of APP in hippocampal neurons and dampens secretion of the inflammatory cytokine tumor necrosis factor-alpha by microglial cells in the familial AD APPswe/PS1ΔE9 (APP/PS1) mouse model. Moreover, APP/PS1 mice with kinase-inactive PI3Kδ (δD910A) had reduced Aß peptides levels and plaques in the brain and an abrogated inflammatory response compared with APP/PS1 littermates. Mechanistic investigations reveal that PI3Kδ inhibition decreases the axonal transport of APP by eliciting the formation of highly elongated tubular-shaped APP-containing carriers, reducing the levels of secreted Aß peptide. Importantly, APP/PS1/δD910A mice exhibited no spatial learning or memory deficits. Our data highlight inhibition of PI3Kδ as a new approach to protect against AD pathology due to its dual action of dampening microglial-dependent neuroinflammation and reducing plaque burden by inhibition of neuronal APP trafficking and processing.SIGNIFICANCE STATEMENT During Alzheimer's disease (AD), the accumulation of the toxic amyloid-ß (Aß) peptide in plaques is associated with a chronic excessive inflammatory response. Uncovering new drug targets that simultaneously reduce both Aß plaque load and neuroinflammation holds therapeutic promise. Using a combination of genetic and pharmacological approaches, we found that the p110δ isoform of phosphoinositide 3-kinase (PI3K) is involved in anterograde trafficking of the amyloid precursor protein in neurons and in the secretion of tumor necrosis factor-alpha from microglial cells. Genetic inactivation of PI3Kδ reduces Aß plaque deposition and abrogates the inflammatory response, resulting in a complete rescue of the life span and spatial memory performance. We conclude that inhibiting PI3Kδ represents a novel therapeutic approach to ameliorate AD pathology by dampening plaque accumulation and microglial-dependent neuroinflammation.


Alzheimer Disease/prevention & control , Amyloid beta-Protein Precursor/metabolism , Class I Phosphatidylinositol 3-Kinases/genetics , Cognitive Dysfunction/genetics , Cognitive Dysfunction/prevention & control , Encephalitis/genetics , Encephalitis/prevention & control , Plaque, Amyloid/genetics , Plaque, Amyloid/prevention & control , Tumor Necrosis Factor-alpha/metabolism , Alzheimer Disease/genetics , Alzheimer Disease/pathology , Animals , Axonal Transport/genetics , Cytokines/metabolism , Female , Male , Maze Learning , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neurons/metabolism , Point Mutation , Primary Cell Culture , Spatial Memory
...