Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Language
Publication year range
1.
Front Cell Dev Biol ; 11: 1165125, 2023.
Article in English | MEDLINE | ID: mdl-37143894

ABSTRACT

Nerve growth factor (NGF) is critical for neuronal physiology during development and adulthood. Despite the well-recognized effect of NGF on neurons, less is known about whether NGF can actually affect other cell types in the central nervous system (CNS). In this work, we show that astrocytes are susceptible to changes in ambient levels of NGF. First, we observe that interfering with NGF signaling in vivo via the constitutive expression of an antiNGF antibody induces astrocytic atrophy. A similar asthenic phenotype is encountered in an uncleavable proNGF transgenic mouse model (TgproNGF#72), effectively increasing the brain proNGF levels. To examine whether this effect on astrocytes is cell-autonomous, we cultured wild-type primary astrocytes in the presence of antiNGF antibodies, uncovering that a short incubation period is sufficient to potently and rapidly trigger calcium oscillations. Acute induction of calcium oscillations by antiNGF antibodies is followed by progressive morphological changes similar to those observed in antiNGF AD11 mice. Conversely, incubation with mature NGF has no effect on either calcium activity nor on astrocytic morphology. At longer timescales, transcriptomic analysis revealed that NGF-deprived astrocytes acquire a proinflammatory profile. In particular, antiNGF-treated astrocytes show upregulation of neurotoxic transcripts and downregulation of neuroprotective mRNAs. Consistent with that data, culturing wild-type neurons in the presence of NGF-deprived astrocytes leads to neuronal cell death. Finally, we report that in both awake and anesthetized mice, astrocytes in layer I of the motor cortex respond with an increase in calcium activity to acute NGF inhibition using either NGF-neutralizing antibodies or a TrkA-Fc NGF scavenger. Moreover, in vivo calcium imaging in the cortex of the 5xFAD neurodegeneration mouse model shows an increased level of spontaneous calcium activity in astrocytes, which is significantly reduced after acute administration of NGF. In conclusion, we unveil a novel neurotoxic mechanism driven by astrocytes, triggered by their sensing and reacting to changes in the levels of ambient NGF.

2.
Glia ; 66(7): 1395-1416, 2018 07.
Article in English | MEDLINE | ID: mdl-29473218

ABSTRACT

Microglia are the sentinels of the brain but a clear understanding of the factors that modulate their activation in physiological and pathological conditions is still lacking. Here we demonstrate that Nerve Growth Factor (NGF) acts on microglia by steering them toward a neuroprotective and anti-inflammatory phenotype. We show that microglial cells express functional NGF receptors in vitro and ex vivo. Our transcriptomic analysis reveals how, in primary microglia, NGF treatment leads to a modulation of motility, phagocytosis and degradation pathways. At the functional level, NGF induces an increase in membrane dynamics and macropinocytosis and, in vivo, it activates an outward rectifying current that appears to modulate glutamatergic neurotransmission in nearby neurons. Since microglia are supposed to be a major player in Aß peptide clearance in the brain, we tested the effects of NGF on its phagocytosis. NGF was shown to promote TrkA-mediated engulfment of Aß by microglia, and to enhance its degradation. Additionally, the proinflammatory activation induced by Aß treatment is counteracted by the concomitant administration of NGF. Moreover, by acting specifically on microglia, NGF protects neurons from the Aß-induced loss of dendritic spines and inhibition of long term potentiation. Finally, in an ex-vivo setup of acute brain slices, we observed a similar increase in Aß engulfment by microglial cells under the influence of NGF. Our work substantiates a role for NGF in the regulation of microglial homeostatic activities and points toward this neurotrophin as a neuroprotective agent in Aß accumulation pathologies, via its anti-inflammatory activity on microglia.


Subject(s)
Microglia/metabolism , Nerve Growth Factor/metabolism , Neuroprotection/physiology , Receptors, Nerve Growth Factor/metabolism , Amyloid beta-Peptides/metabolism , Animals , Brain/cytology , Brain/metabolism , CX3C Chemokine Receptor 1/genetics , CX3C Chemokine Receptor 1/metabolism , Cells, Cultured , Coculture Techniques , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Male , Mice, Inbred C57BL , Mice, Transgenic , Microglia/cytology , Nerve Growth Factor/administration & dosage , Neurons/cytology , Neurons/metabolism , Phagocytosis/physiology , Receptors, Nerve Growth Factor/antagonists & inhibitors , Synaptic Transmission/physiology , Tissue Culture Techniques , Transcriptome
3.
Brain ; 140(1): 201-217, 2017 01.
Article in English | MEDLINE | ID: mdl-28031222

ABSTRACT

Nerve growth factor is a therapeutic candidate for Alzheimer's disease. Due to its pain-inducing activity, in current clinical trials nerve growth factor is delivered locally into the brain by neurosurgery, but data on the efficacy of local nerve growth factor delivery in decreasing amyloid-ß deposition are not available. To reduce the nerve growth factor pain-inducing side effects, thus avoiding the need for local brain injection, we developed human painless nerve growth factor (hNGFp), inspired by the human genetic disease hereditary sensory and autonomic neuropathy type V. hNGFp has identical neurotrophic potency as wild-type human nerve growth factor, but a 10-fold lower pain sensitizing activity. In this study we first mimicked, in the 5xFAD mouse model, the intraparenchymal delivery of hNGFp used in clinical trials and found it to be ineffective in decreasing amyloid-ß plaque load. On the contrary, the same dose of hNGFp delivered intranasally, which was widely biodistributed in the brain and did not induce pain, showed a potent anti-amyloidogenic action and rescued synaptic plasticity and memory deficits. We found that hNGFp acts on glial cells, modulating inflammatory proteins such as the soluble TNFα receptor II and the chemokine CXCL12. We further established that the rescuing effect by hNGFp is mediated by CXCL12, as pharmacological inhibition of CXCL12 receptor CXCR4 occludes most of hNGFp effects. These findings have significant therapeutic implications: (i) we established that a widespread exposure of the brain is required for nerve growth factor to fully exert its neuroprotective actions; and (ii) we have identified a new anti-neurodegenerative pathway as a broad target for new therapeutic opportunities for neurodegenerative diseases.


Subject(s)
Alzheimer Disease/drug therapy , Cerebral Cortex/metabolism , Chemokine CXCL12/metabolism , Memory Disorders/drug therapy , Nerve Growth Factor/pharmacology , Neuronal Plasticity/drug effects , Pain/chemically induced , Plaque, Amyloid/drug therapy , Administration, Intranasal , Animals , Behavior, Animal , Cerebral Cortex/drug effects , Disease Models, Animal , Humans , Mice , Mice, Transgenic , Nerve Growth Factor/administration & dosage , Nerve Growth Factor/adverse effects , Receptors, CXCR4/antagonists & inhibitors
4.
J Alzheimers Dis ; 42(1): 103-7, 2014.
Article in English | MEDLINE | ID: mdl-24844685

ABSTRACT

Autosomal dominant forms of familial Alzheimer's disease are linked to an aberrant processing of the amyloid-ß protein precursor, which results in an increased production of amyloid-ß (Aß) peptides that first form oligomers and eventually aggregate in the form of extracellular amyloid plaques in the brain. The accumulation of Aß peptides oligomers seems to correlate with alterations of synaptic transmission in experimental models of Alzheimer's disease. Whether Aß aggregation disrupts synaptic function independently of amyloid plaques deposition still needs further research. Here we report an amyloid plaque-independent deficit of neuronal plasticity after short-term sensory deprivation in the visual system of 5XFAD mice.


Subject(s)
Alzheimer Disease/physiopathology , Neuronal Plasticity/physiology , Sensory Deprivation/physiology , Visual Cortex/physiopathology , Visual Perception/physiology , Animals , Disease Models, Animal , Mice , Mice, Transgenic , Plaque, Amyloid/physiopathology , Time Factors , Vision, Binocular/physiology , Vision, Monocular/physiology
5.
J Alzheimers Dis ; 31(1): 1-6, 2012.
Article in English | MEDLINE | ID: mdl-22504318

ABSTRACT

Several studies suggest that systemic infection occurring during aging and chronic neurodegenerative diseases can evoke an exaggerated immune response that contributes to the progression of neurodegeneration and cognitive decline. However, studies directly addressing the relationship between microbial environment and the onset of neurodegeneration in Alzheimer's disease animal models are lacking. Here we show that the onset of neurodegeneration that transgenic mice develop when raised in conventional husbandry slows down when raising anti-nerve growth factor transgenic mice in a murine pathogen free condition.


Subject(s)
Nerve Growth Factor/deficiency , Neurodegenerative Diseases/complications , Neurodegenerative Diseases/etiology , Animals , Caliciviridae Infections/complications , Choline O-Acetyltransferase/metabolism , Cognition Disorders/etiology , Disease Models, Animal , Disease Progression , Humans , Interleukin-6/blood , Lipopolysaccharides/toxicity , Mice , Mice, Transgenic , Neurodegenerative Diseases/blood , Neurodegenerative Diseases/pathology , Norovirus/pathogenicity , Trichomonas Infections/complications , tau Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...