Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
Add more filters










Publication year range
1.
Neurophotonics ; 11(1): 014415, 2024 Jan.
Article in English | MEDLINE | ID: mdl-38545127

ABSTRACT

The Frontiers in Neurophotonics Symposium is a biennial event that brings together neurobiologists and physicists/engineers who share interest in the development of leading-edge photonics-based approaches to understand and manipulate the nervous system, from its individual molecular components to complex networks in the intact brain. In this Community paper, we highlight several topics that have been featured at the symposium that took place in October 2022 in Québec City, Canada.

2.
bioRxiv ; 2023 Sep 23.
Article in English | MEDLINE | ID: mdl-37790349

ABSTRACT

Women are the main target of intimate partner violence (IPV), which is escalating worldwide. Mechanisms subtending IPV-related disorders, such as anxiety, depression and PTSD, remain unclear. We employed a mouse model molded on an IPV scenario (male vs. female prolonged violent interaction) to unearth the neuroendocrine alterations triggered by an aggressive male mouse on the female murine brain. Experimental IPV (EIPV) prompted marked anxiety-like behavior in young female mice, coincident with high circulating/cerebral corticosterone levels. The hippocampus of EIPV-inflicted female animals displayed neuronal loss, reduced BrdU-DCX-positive nuclei, decreased mature DCX-positive cells, and diminished dendritic arborization level in the dentate gyrus (DG), features denoting impaired neurogenesis and neuronal differentiation. These hallmarks were associated with marked down-regulation of estrogen receptor ß (ERß) density in the hippocampus, especially in the DG and dependent prosurvival ERK signaling. Conversely, ERα expression was unchanged. After EIPV, the DG harbored lowered local BDNF pools, diminished TrkB phosphorylation, and elevated glucocorticoid receptor phosphorylation. In unison, ERß KO mice had heightened anxiety-like behavior and curtailed BDNF levels at baseline, despite enhanced circulating estradiol levels, while dying prematurely during EIPV. Thus, reiterated male-to-female violence jeopardizes hippocampal homeostasis in the female brain, perturbing ERß/BDNF signaling, thus instigating anxiety and chronic stress.

3.
iScience ; 26(1): 105728, 2023 Jan 20.
Article in English | MEDLINE | ID: mdl-36582822

ABSTRACT

In Neurodevelopmental Disorders, alterations of synaptic plasticity may trigger structural changes in neuronal circuits involved in cognitive functions. This hypothesis was tested in mice carrying the human R451C mutation of Nlgn3 gene (NLG3R451C KI), found in some families with autistic children. To this aim, the spike time dependent plasticity (STDP) protocol was applied to immature GABAergic Mossy Fibers (MF)-CA3 connections in hippocampal slices from NLG3R451C KI mice. These animals failed to exhibit STD-LTP, an effect that persisted in adulthood when these synapses became glutamatergic. Similar results were obtained in mice lacking the Nlgn3 gene (NLG3 KO mice), suggesting a loss of function. The loss of STD-LTP was associated with a premature shift of GABA from the depolarizing to the hyperpolarizing direction, a reduced BDNF availability and TrkB phosphorylation at potentiated synapses. These effects may constitute a general mechanism underlying cognitive deficits in those forms of Autism caused by synaptic dysfunctions.

4.
Cells ; 11(9)2022 04 29.
Article in English | MEDLINE | ID: mdl-35563806

ABSTRACT

Neurons release and respond to brain-derived neurotrophic factor (BDNF) with bursts of brain activity. BDNF action is known to extend to peri-synaptic astrocytes, contributing to synaptic strengthening. This implies that astrocytes have a set of dynamic responses, some of which might be secondary to activation of the tropomyosin tyrosine kinase B (TrkB) receptor. Here, we assessed the contribution of BDNF to long-term synaptic potentiation (LTP), by specifically deleting TrkB in cortical astrocytes. TrkB deletion had no effect on LTP induction, stabilization and maintenance, indicating that TrkB signaling in astrocytes is extraneous to transducing BDNF activity for synaptic strengthening.


Subject(s)
Brain-Derived Neurotrophic Factor , Perirhinal Cortex , Astrocytes/metabolism , Brain-Derived Neurotrophic Factor/metabolism , Perirhinal Cortex/metabolism , Protein-Tyrosine Kinases/metabolism , Receptor, trkB/metabolism
5.
Biomolecules ; 12(2)2022 01 23.
Article in English | MEDLINE | ID: mdl-35204694

ABSTRACT

Metastatic prostate cancer (mPCa) is one of the leading causes of cancer-related mortality in both the US and Europe. Androgen deprivation is the first-line therapy for mPCa; however, resistance to therapy inevitably occurs and the disease progresses to the castration resistant stage, which is uncurable. A definition of novel targeted therapies is necessary for the establishment of innovative and more effective protocols of personalized oncology. We employed genetically engineered mouse models of PCa and human samples to characterize the expression of the TRPM8 cation channel in both hormone naïve and castration resistant tumors. We show that Trpm8 expression marks both indolent (Pten-null) and aggressive (Pten/Trp53 double-null and TRAMP) mouse prostate adenocarcinomas. Importantly, both mouse and human castration-resistant PCa preserve TRPM8 protein expression. Finally, we tested the effect of TRPM8 agonist D-3263 administration in combination with enzalutamide or docetaxel on the viability of aggressive mouse PCa cell lines. Our data demonstrate that D-3263 substantially enhances the pro-apoptotic activity of enzalutamide and docetaxel in TRAMP-C1 e TRAMP-C2 PCa cell lines. To conclude, this study provides the basis for pre-clinical in vivo testing of TRPM8 targeting as a novel strategy to implement the efficacy of standard-of-care treatments for advanced PCa.


Subject(s)
Adenocarcinoma , Membrane Proteins , Prostatic Neoplasms, Castration-Resistant , TRPM Cation Channels , Adenocarcinoma/drug therapy , Adenocarcinoma/metabolism , Androgen Antagonists/therapeutic use , Animals , Castration , Humans , Male , Membrane Proteins/metabolism , Mice , Prostatic Neoplasms, Castration-Resistant/drug therapy , Prostatic Neoplasms, Castration-Resistant/metabolism
6.
Commun Biol ; 4(1): 1152, 2021 10 05.
Article in English | MEDLINE | ID: mdl-34611268

ABSTRACT

Memory consolidation requires astrocytic microdomains for protein recycling; but whether this lays a mechanistic foundation for long-term information storage remains enigmatic. Here we demonstrate that persistent synaptic strengthening invited astrocytic microdomains to convert initially internalized (pro)-brain-derived neurotrophic factor (proBDNF) into active prodomain (BDNFpro) and mature BDNF (mBDNF) for synaptic re-use. While mBDNF activates TrkB, we uncovered a previously unsuspected function for the cleaved BDNFpro, which increases TrkB/SorCS2 receptor complex at post-synaptic sites. Astrocytic BDNFpro release reinforced TrkB phosphorylation to sustain long-term synaptic potentiation and to retain memory in the novel object recognition behavioral test. Thus, the switch from one inactive state to a multi-functional one of the proBDNF provides post-synaptic changes that survive the initial activation. This molecular asset confines local information storage in astrocytic microdomains to selectively support memory circuits.


Subject(s)
Astrocytes/physiology , Brain-Derived Neurotrophic Factor/genetics , Long-Term Potentiation/genetics , Membrane Glycoproteins/genetics , Memory/physiology , Nerve Tissue Proteins/genetics , Protein-Tyrosine Kinases/genetics , Receptors, Cell Surface/genetics , Animals , Brain-Derived Neurotrophic Factor/metabolism , Membrane Glycoproteins/metabolism , Mice , Nerve Tissue Proteins/metabolism , Protein-Tyrosine Kinases/metabolism , Receptors, Cell Surface/metabolism
7.
Commun Biol ; 4(1): 62, 2021 01 12.
Article in English | MEDLINE | ID: mdl-33437023

ABSTRACT

Recent computational advancements in the simulation of biochemical processes allow investigating the mechanisms involved in protein regulation with realistic physics-based models, at an atomistic level of resolution. These techniques allowed us to design a drug discovery approach, named Pharmacological Protein Inactivation by Folding Intermediate Targeting (PPI-FIT), based on the rationale of negatively regulating protein levels by targeting folding intermediates. Here, PPI-FIT was tested for the first time on the cellular prion protein (PrP), a cell surface glycoprotein playing a key role in fatal and transmissible neurodegenerative pathologies known as prion diseases. We predicted the all-atom structure of an intermediate appearing along the folding pathway of PrP and identified four different small molecule ligands for this conformer, all capable of selectively lowering the load of the protein by promoting its degradation. Our data support the notion that the level of target proteins could be modulated by acting on their folding pathways, implying a previously unappreciated role for folding intermediates in the biological regulation of protein expression.


Subject(s)
Drug Evaluation, Preclinical/methods , Prion Diseases/drug therapy , Prion Proteins/chemistry , Prion Proteins/metabolism , Protein Folding , Animals , Binding Sites , Computer Simulation , Endoplasmic Reticulum/metabolism , Fibroblasts , HEK293 Cells , Humans , Ligands , Lysosomes/drug effects , Lysosomes/metabolism , Mice , Peptide Fragments/chemistry , Peptide Fragments/metabolism , Protein Processing, Post-Translational , Reproducibility of Results
8.
Hum Mol Genet ; 26(19): 3749-3762, 2017 10 01.
Article in English | MEDLINE | ID: mdl-28934387

ABSTRACT

Spinocerebellar ataxia type 35 (SCA35) is a rare autosomal-dominant neurodegenerative disease caused by mutations in the TGM6 gene, which codes for transglutaminase 6 (TG6). Mutations in TG6 induce cerebellar degeneration by an unknown mechanism. We identified seven patients bearing new mutations in TGM6. To gain insights into the molecular basis of mutant TG6-induced neurotoxicity, we analyzed all the seven new TG6 mutants and the five TG6 mutants previously linked to SCA35. We found that the wild-type (TG6-WT) protein mainly localized to the nucleus and perinuclear area, whereas five TG6 mutations showed nuclear depletion, increased accumulation in the perinuclear area, insolubility and loss of enzymatic function. Aberrant accumulation of these TG6 mutants in the perinuclear area led to activation of the unfolded protein response (UPR), suggesting that specific TG6 mutants elicit an endoplasmic reticulum stress response. Mutations associated with activation of the UPR caused death of primary neurons and reduced the survival of novel Drosophila melanogaster models of SCA35. These results indicate that mutations differently impacting on TG6 function cause neuronal dysfunction and death through diverse mechanisms and highlight the UPR as a potential therapeutic target for patient treatment.


Subject(s)
Spinocerebellar Ataxias/genetics , Transglutaminases/genetics , Transglutaminases/metabolism , Unfolded Protein Response/genetics , Animals , Animals, Genetically Modified , COS Cells , Cell Line , Chlorocebus aethiops , Drosophila melanogaster , Endoplasmic Reticulum/metabolism , Endoplasmic Reticulum Stress/genetics , Female , HEK293 Cells , Humans , Male , Mice, Inbred C57BL , Mutation , Neurons/enzymology , Neurons/metabolism , Neurons/pathology , Spinocerebellar Ataxias/enzymology , Spinocerebellar Ataxias/metabolism , Spinocerebellar Ataxias/pathology
10.
Pflugers Arch ; 469(5-6): 593-610, 2017 06.
Article in English | MEDLINE | ID: mdl-28280960

ABSTRACT

Brain-derived neurotrophic factor (BDNF) is a member of the neurotrophin family of secreted proteins. Signaling cascades induced by BDNF and its receptor, the receptor tyrosine kinase TrkB, link neuronal growth and differentiation with synaptic plasticity. For this reason, interference with BDNF signaling has emerged as a promising strategy for potential treatments in psychiatric and neurological disorders. In many brain circuits, synaptically released BDNF is essential for structural and functional long-term potentiation, two prototypical cellular models of learning and memory formation. Recent studies have revealed an unexpected complexity in the synaptic communication of mature BDNF and its precursor proBDNF, not only between local pre- and postsynaptic neuronal targets but also with participation of glial cells. Here, we consider recent findings on local actions of the BDNF family of ligands at the synapse and discuss converging lines of evidence which emerge from per se conflicting results.


Subject(s)
Brain-Derived Neurotrophic Factor/metabolism , Synapses/metabolism , Animals , Anxiety Disorders/metabolism , Brain-Derived Neurotrophic Factor/genetics , Humans , Long-Term Potentiation , Receptor, trkB/metabolism , Synapses/physiology , Synaptic Transmission
11.
Commun Integr Biol ; 10(1): e1277296, 2017.
Article in English | MEDLINE | ID: mdl-28289489

ABSTRACT

We have recently reported that long-term memory retention requires synaptic glia for proBDNF uptake and recycling. Through the recycling course, glial cells release endocytic BDNF, a mechanism that is activated in response to glutamate via AMPA and mGluRI/II receptors. Cortical astrocytes express receptors for many different transmitters suggesting for a complex signaling controlling endocytic BDNF secretion. Here, we demonstrated that the extracellular nucleotide ATP, activating P2X and P2Y receptors, regulates endocytic BDNF secretion in cultured astrocytes. Our data indicate that distinct glioactive molecules can participate in BDNF glial recycling and suggest that cortical astrocytes contributing to neuronal plasticity can be influenced by neurotransmitters in tune with synaptic needs.

12.
Neurobiol Dis ; 100: 75-86, 2017 Apr.
Article in English | MEDLINE | ID: mdl-28088401

ABSTRACT

Oligophrenin-1 (OPHN1) is a Rho GTPase activating protein whose mutations cause X-linked intellectual disability (XLID). How loss of function of Ophn1 affects neuronal development is only partly understood. Here we have exploited adult hippocampal neurogenesis to dissect the steps of neuronal differentiation that are affected by Ophn1 deletion. We found that mice lacking Ophn1 display a reduction in the number of newborn neurons in the dentate gyrus. A significant fraction of the Ophn1-deficient newly generated neurons failed to extend an axon towards CA3, and showed an altered density of dendritic protrusions. Since Ophn1-deficient mice display overactivation of Rho-associated protein kinase (ROCK) and protein kinase A (PKA) signaling, we administered a clinically approved ROCK/PKA inhibitor (fasudil) to correct the neurogenesis defects. While administration of fasudil was not effective in rescuing axon formation, the same treatment completely restored spine density to control levels, and enhanced the long-term survival of adult-born neurons in mice lacking Ophn1. These results identify specific neurodevelopmental steps that are impacted by Ophn1 deletion, and indicate that they may be at least partially corrected by pharmacological treatment.


Subject(s)
Hippocampus/metabolism , Intellectual Disability/physiopathology , Neurogenesis/physiology , Neurons/metabolism , Animals , Cytoskeletal Proteins/deficiency , Cytoskeletal Proteins/metabolism , Disease Models, Animal , GTPase-Activating Proteins/deficiency , GTPase-Activating Proteins/metabolism , Mice, Inbred C57BL , Mice, Knockout , Nuclear Proteins/deficiency , Nuclear Proteins/metabolism
13.
Neuron ; 92(4): 873-887, 2016 Nov 23.
Article in English | MEDLINE | ID: mdl-27746130

ABSTRACT

Glial cells respond to neuronal activation and release neuroactive molecules (termed "gliotransmitters") that can affect synaptic activity and modulate plasticity. In this study, we used molecular genetic tools, ultra-structural microscopy, and electrophysiology to assess the role of brain-derived neurotrophic factor (BDNF) on cortical gliotransmission in vivo. We find that glial cells recycle BDNF that was previously secreted by neurons as pro-neurotrophin following long-term potentiation (LTP)-inducing electrical stimulation. Upon BDNF glial recycling, we observed tight, temporal, highly localized TrkB phosphorylation on adjacent neurons, a process required to sustain LTP. Engagement of BDNF recycling by astrocytes represents a novel mechanism by which cortical synapses can expand BDNF action and provide synaptic changes that are relevant for the acquisition of new memories. Accordingly, mice deficient in BDNF glial recycling fail to recognize familiar from novel objects, indicating a physiological requirement for this process in memory consolidation.


Subject(s)
Brain-Derived Neurotrophic Factor/metabolism , Long-Term Potentiation , Memory , Neuroglia/metabolism , Receptor, trkB/metabolism , Receptors, Nerve Growth Factor/metabolism , Synapses/metabolism , Animals , Cerebral Cortex/cytology , Cerebral Cortex/metabolism , Mice , Phosphorylation , Receptors, Nerve Growth Factor/genetics
14.
Cell Rep ; 7(1): 138-52, 2014 Apr 10.
Article in English | MEDLINE | ID: mdl-24685135

ABSTRACT

VIDEO ABSTRACT: Newly generated neurons initiate polarizing signals that specify a single axon and multiple dendrites, a process critical for patterning neuronal circuits in vivo. Here, we report that the pan-neurotrophin receptor p75(NTR) is a polarity regulator that localizes asymmetrically in differentiating neurons in response to neurotrophins and is required for specification of the future axon. In cultured hippocampal neurons, local exposure to neurotrophins causes early accumulation of p75(NTR) into one undifferentiated neurite to specify axon fate. Moreover, knockout or knockdown of p75(NTR) results in failure to initiate an axon in newborn neurons upon cell-cycle exit in vitro and in the developing cortex, as well as during adult hippocampal neurogenesis in vivo. Hence, p75(NTR) governs neuronal polarity, determining pattern and assembly of neuronal circuits in adult hippocampus and cortical development.


Subject(s)
Axons/metabolism , Neurons/metabolism , Receptor, Nerve Growth Factor/metabolism , Animals , Cell Polarity/physiology , Cells, Cultured , Gene Knockdown Techniques , Hippocampus/cytology , Hippocampus/metabolism , Mice , Mice, Knockout , Neurogenesis , Neurons/cytology , Stem Cells/metabolism
15.
J Neurosci ; 33(28): 11464-78, 2013 Jul 10.
Article in English | MEDLINE | ID: mdl-23843518

ABSTRACT

In the adult rodent brain, the olfactory bulb (OB) is continuously supplied with new neurons which survival critically depends on their successful integration into pre-existing networks. Yet, the extracellular signals that determine the selection which neurons will be ultimately incorporated into these circuits are largely unknown. Here, we show that immature neurons express the catalytic form of the brain-derived neurotrophic factor receptor TrkB [full-length TrkB (TrkB-FL)] only after their arrival in the OB, at the time when integration commences. To unravel the role of TrkB signaling in newborn neurons, we conditionally ablated TrkB-FL in mice via Cre expression in adult neural stem and progenitor cells. TrkB-deficient neurons displayed a marked impairment in dendritic arborization and spine growth. By selectively manipulating the signaling pathways initiated by TrkB in vivo, we identified the transducers Shc/PI3K to be required for dendritic growth, whereas the activation of phospholipase C-γ was found to be responsible for spine formation. Furthermore, long-term genetic fate mapping revealed that TrkB deletion severely compromised the survival of new dopaminergic neurons, leading to a substantial reduction in the overall number of adult-generated periglomerular cells (PGCs), but not of granule cells (GCs). Surprisingly, this loss of dopaminergic PGCs was mirrored by a corresponding increase in the number of calretinin+ PGCs, suggesting that distinct subsets of adult-born PGCs may respond differentially to common extracellular signals. Thus, our results identify TrkB signaling to be essential for balancing the incorporation of defined classes of adult-born PGCs and not GCs, reflecting their different mode of integration in the OB.


Subject(s)
Adult Stem Cells/physiology , Neural Stem Cells/physiology , Neurogenesis/physiology , Olfactory Bulb/cytology , Olfactory Bulb/growth & development , Receptor, trkB/physiology , Signal Transduction/physiology , Age Factors , Animals , Animals, Newborn , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Receptor, trkB/deficiency , Receptor, trkB/genetics
16.
Cell Rep ; 3(6): 1824-31, 2013 Jun 27.
Article in English | MEDLINE | ID: mdl-23770243

ABSTRACT

Immunoglobulin (Ig) isotype diversification by class switch recombination (CSR) is an essential process for mounting a protective humoral immune response. Ig CSR deficiencies in humans can result from an intrinsic B cell defect; however, most of these deficiencies are still molecularly undefined and diagnosed as common variable immunodeficiency (CVID). Here, we show that extracellular adenosine critically contributes to CSR in human naive and IgM memory B cells. In these cells, coordinate stimulation of B cell receptor and toll-like receptors results in the release of ATP stored in Ca(2+)-sensitive secretory vesicles. Plasma membrane ectonucleoside triphosphate diphosphohydrolase 1 CD39 and ecto-5'-nucleotidase CD73 hydrolyze ATP to adenosine, which induces CSR in B cells in an autonomous fashion. Notably, CVID patients with impaired class-switched antibody responses are selectively deficient in CD73 expression in B cells, suggesting that CD73-dependent adenosine generation contributes to the pathogenesis of this disease.


Subject(s)
5'-Nucleotidase/immunology , Adenosine Triphosphate/immunology , Antibody Formation/immunology , B-Lymphocytes/immunology , Immunoglobulin Class Switching/immunology , 5'-Nucleotidase/genetics , 5'-Nucleotidase/metabolism , Adenosine Triphosphate/genetics , Adenosine Triphosphate/metabolism , Animals , Antibody Formation/genetics , Antigens, CD/immunology , Antigens, CD/metabolism , Apyrase/immunology , Apyrase/metabolism , B-Lymphocyte Subsets/cytology , B-Lymphocyte Subsets/immunology , B-Lymphocytes/cytology , B-Lymphocytes/metabolism , Common Variable Immunodeficiency/genetics , Common Variable Immunodeficiency/immunology , Common Variable Immunodeficiency/metabolism , Humans , Mice , Mice, Transgenic , Recombination, Genetic
17.
Biol Cell ; 104(4): 213-28, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22188132

ABSTRACT

BACKGROUND INFORMATION: ATP is the main transmitter stored and released from astrocytes under physiological and pathological conditions. Morphological and functional evidence suggest that besides secretory granules, secretory lysosomes release ATP. However, the molecular mechanisms involved in astrocytic lysosome fusion remain still unknown. RESULTS: In the present study, we identify tetanus neurotoxin-insensitive vesicle-associated membrane protein (TI-VAMP, also called VAMP7) as the vesicular SNARE which mediates secretory lysosome exocytosis, contributing to release of both ATP and cathepsin B from glial cells. We also demonstrate that fusion of secretory lysosomes is triggered by slow and locally restricted calcium elevations, distinct from calcium spikes which induce the fusion of glutamate-containing clear vesicles. Downregulation of TI-VAMP/VAMP7 expression inhibited the fusion of ATP-storing vesicles and ATP-mediated calcium wave propagation. TI-VAMP/VAMP7 downregulation also significantly reduced secretion of cathepsin B from glioma. CONCLUSIONS: Given that sustained ATP release from glia upon injury greatly contributes to secondary brain damage and cathepsin B plays a critical role in glioma dissemination, TI-VAMP silencing can represent a novel strategy to control lysosome fusion in pathological conditions.


Subject(s)
Adenosine Triphosphate/metabolism , Astrocytes/metabolism , Calcium/metabolism , Cathepsin B/metabolism , Lysosomes/metabolism , R-SNARE Proteins/metabolism , Animals , Astrocytes/cytology , Cerebral Cortex/cytology , Cerebral Cortex/embryology , Cerebral Cortex/metabolism , Down-Regulation , Embryo, Mammalian , Exocytosis , Glioma/metabolism , Glioma/pathology , Hippocampus/cytology , Hippocampus/embryology , Hippocampus/metabolism , Humans , Membrane Fusion , Neuroglia/cytology , Neuroglia/metabolism , Primary Cell Culture , Protein Binding , R-SNARE Proteins/antagonists & inhibitors , R-SNARE Proteins/genetics , RNA, Small Interfering/genetics , Rats , Signal Transduction , Transfection
18.
Commun Integr Biol ; 2(1): 14-6, 2009.
Article in English | MEDLINE | ID: mdl-19704856

ABSTRACT

Brain-derived neurotrophic factor (BDNF) is a member of the neurotrophin family, which has been reported to regulate neurogenesis in the dentate gyrus, but the molecular control over this process remains unclear. We demonstrated that by activating TrkB receptor tyrosine kinase, BDNF controls the size of the surviving pool of newborn neurons at the time of connectivity. The TrkB-dependent decision regarding survival in these newborn neurons takes place at approximately four to six weeks of age. Before newborn neurons start to die they exhibit a drastic reduction in dendritic complexity and spine density, which may reflect a failure of these cells to integrate appropriately. Both the failure to become integrated, and subsequent dying, leads to impaired neurogenesisdependent plasticity and increased anxiety-like behavior in mice lacking a functional TrkB receptor in newborn neurons. Thus, our data demonstrate the importance of BDNF/TrkB signaling for the survival and integration of newborn neurons in the adult hippocampus and suggest a critical function of these neurons in regulating the anxiety state of the animal.

19.
J Cell Biol ; 183(2): 213-21, 2008 Oct 20.
Article in English | MEDLINE | ID: mdl-18852301

ABSTRACT

Activity-dependent secretion of brain-derived neurotrophic factor (BDNF) is thought to enhance synaptic plasticity, but the mechanisms controlling extracellular availability and clearance of secreted BDNF are poorly understood. We show that BDNF is secreted in its precursor form (pro-BDNF) and is then cleared from the extracellular space through rapid uptake by nearby astrocytes after theta-burst stimulation in layer II/III of cortical slices, a paradigm resulting in long-term potentiation of synaptic transmission. Internalization of pro-BDNF occurs via the formation of a complex with the pan-neurotrophin receptor p75 and subsequent clathrin-dependent endocytosis. Fluorescence-tagged pro-BDNF and real-time total internal reflection fluorescence microscopy in cultured astrocytes is used to monitor single endocytic vesicles in response to the neurotransmitter glutamate. We find that endocytosed pro-BDNF is routed into a fast recycling pathway for subsequent soluble NSF attachment protein receptor-dependent secretion. Thus, astrocytes contain an endocytic compartment competent for pro-BDNF recycling, suggesting a specialized form of bidirectional communication between neurons and glia.


Subject(s)
Astrocytes/cytology , Astrocytes/metabolism , Brain-Derived Neurotrophic Factor/metabolism , Cerebral Cortex/cytology , Endocytosis , Neurotransmitter Agents/metabolism , Protein Precursors/metabolism , Animals , Astrocytes/ultrastructure , Cells, Cultured , Clathrin/metabolism , Mice , Neurons/cytology , Neurons/metabolism , Rats , Rats, Wistar , Receptor, Nerve Growth Factor/metabolism , Synaptic Transmission , Synaptic Vesicles/metabolism , Vesicle-Associated Membrane Protein 2/metabolism
20.
Proc Natl Acad Sci U S A ; 105(40): 15570-5, 2008 Oct 07.
Article in English | MEDLINE | ID: mdl-18832146

ABSTRACT

New neurons in the adult dentate gyrus are widely held to incorporate into hippocampal circuitry via a stereotypical sequence of morphological and physiological transitions, yet the molecular control over this process remains unclear. We studied the role of brain-derived neurotrophic factor (BDNF)/TrkB signaling in adult neurogenesis by deleting the full-length TrkB via Cre expression within adult progenitors in TrkB(lox/lox) mice. By 4 weeks after deletion, the growth of dendrites and spines is reduced in adult-born neurons demonstrating that TrkB is required to create the basic organization of synaptic connections. Later, when new neurons normally display facilitated synaptic plasticity and become preferentially recruited into functional networks, lack of TrkB results in impaired neurogenesis-dependent long-term potentiation and cell survival becomes compromised. Because of the specific lack of TrkB signaling in recently generated neurons a remarkably increased anxiety-like behavior was observed in mice carrying the mutation, emphasizing the contribution of adult neurogenesis in regulating mood-related behavior.


Subject(s)
Anxiety/genetics , Hippocampus/physiology , Neurons/physiology , Receptor, trkB/genetics , Stem Cells/physiology , Animals , Anxiety/metabolism , Brain-Derived Neurotrophic Factor/metabolism , Dendrites/metabolism , Dentate Gyrus/metabolism , Hippocampus/metabolism , Hippocampus/ultrastructure , Mice , Mice, Transgenic , Mutation , Neuronal Plasticity , Neurons/metabolism , Neurons/ultrastructure , Receptor, trkB/metabolism , Stem Cells/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...