Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 61
Filter
Add more filters










Publication year range
1.
Cancer Discov ; 14(4): 669-673, 2024 Apr 04.
Article in English | MEDLINE | ID: mdl-38571430

ABSTRACT

SUMMARY: The field of cancer neuroscience has begun to define the contributions of nerves to cancer initiation and progression; here, we highlight the future directions of basic and translational cancer neuroscience for malignancies arising outside of the central nervous system.


Subject(s)
Neoplasms , Neurosciences , Humans , Central Nervous System , Forecasting , Proteomics
2.
Cancer Cell ; 42(3): 474-486.e12, 2024 Mar 11.
Article in English | MEDLINE | ID: mdl-38402610

ABSTRACT

Chronic stress is associated with increased risk of metastasis and poor survival in cancer patients, yet the reasons are unclear. We show that chronic stress increases lung metastasis from disseminated cancer cells 2- to 4-fold in mice. Chronic stress significantly alters the lung microenvironment, with fibronectin accumulation, reduced T cell infiltration, and increased neutrophil infiltration. Depleting neutrophils abolishes stress-induced metastasis. Chronic stress shifts normal circadian rhythm of neutrophils and causes increased neutrophil extracellular trap (NET) formation via glucocorticoid release. In mice with neutrophil-specific glucocorticoid receptor deletion, chronic stress fails to increase NETs and metastasis. Furthermore, digesting NETs with DNase I prevents chronic stress-induced metastasis. Together, our data show that glucocorticoids released during chronic stress cause NET formation and establish a metastasis-promoting microenvironment. Therefore, NETs could be targets for preventing metastatic recurrence in cancer patients, many of whom will experience chronic stress due to their disease.


Subject(s)
Extracellular Traps , Lung Neoplasms , Humans , Animals , Mice , Neutrophils/pathology , Lung Neoplasms/pathology , Lung/pathology , Tumor Microenvironment
3.
Nat Commun ; 14(1): 6797, 2023 10 26.
Article in English | MEDLINE | ID: mdl-37884508

ABSTRACT

Axon initial segment (AIS) cell surface proteins mediate key biological processes in neurons including action potential initiation and axo-axonic synapse formation. However, few AIS cell surface proteins have been identified. Here, we use antibody-directed proximity biotinylation to define the cell surface proteins in close proximity to the AIS cell adhesion molecule Neurofascin. To determine the distributions of the identified proteins, we use CRISPR-mediated genome editing for insertion of epitope tags in the endogenous proteins. We identify Contactin-1 (Cntn1) as an AIS cell surface protein. Cntn1 is enriched at the AIS through interactions with Neurofascin and NrCAM. We further show that Cntn1 contributes to assembly of the AIS extracellular matrix, and regulates AIS axo-axonic innervation by inhibitory basket cells in the cerebellum and inhibitory chandelier cells in the cortex.


Subject(s)
Axon Initial Segment , Biological Phenomena , Axon Initial Segment/metabolism , Contactin 1/metabolism , Biotinylation , Synapses/metabolism , Axons/metabolism , Membrane Proteins/metabolism , Antibodies/metabolism
4.
Genes Dev ; 37(15-16): 681-702, 2023 08 01.
Article in English | MEDLINE | ID: mdl-37648371

ABSTRACT

The different cell types in the brain have highly specialized roles with unique metabolic requirements. Normal brain function requires the coordinated partitioning of metabolic pathways between these cells, such as in the neuron-astrocyte glutamate-glutamine cycle. An emerging theme in glioblastoma (GBM) biology is that malignant cells integrate into or "hijack" brain metabolism, co-opting neurons and glia for the supply of nutrients and recycling of waste products. Moreover, GBM cells communicate via signaling metabolites in the tumor microenvironment to promote tumor growth and induce immune suppression. Recent findings in this field point toward new therapeutic strategies to target the metabolic exchange processes that fuel tumorigenesis and suppress the anticancer immune response in GBM. Here, we provide an overview of the intercellular division of metabolic labor that occurs in both the normal brain and the GBM tumor microenvironment and then discuss the implications of these interactions for GBM therapy.


Subject(s)
Glioblastoma , Humans , Brain , Neuroglia , Astrocytes , Neurons , Tumor Microenvironment
5.
bioRxiv ; 2023 Mar 06.
Article in English | MEDLINE | ID: mdl-36945454

ABSTRACT

Axon initial segment (AIS) cell surface proteins mediate key biological processes in neurons including action potential initiation and axo-axonic synapse formation. However, few AIS cell surface proteins have been identified. Here, we used antibody-directed proximity biotinylation to define the cell surface proteins in close proximity to the AIS cell adhesion molecule Neurofascin. To determine the distributions of the identified proteins, we used CRISPR-mediated genome editing for insertion of epitope tags in the endogenous proteins. We found Contactin-1 (Cntn1) among the previously unknown AIS proteins we identified. Cntn1 is enriched at the AIS through interactions with Neurofascin and NrCAM. We further show that Cntn1 contributes to assembly of the AIS-extracellular matrix, and is required for AIS axo-axonic innervation by inhibitory basket cells in the cerebellum and inhibitory chandelier cells in the cortex.

6.
Proc Natl Acad Sci U S A ; 119(11): e2114476119, 2022 03 15.
Article in English | MEDLINE | ID: mdl-35263225

ABSTRACT

SignificanceChandelier cells (ChCs) are a unique type of GABAergic interneuron that form axo-axonic synapses exclusively on the axon initial segment (AIS) of neocortical pyramidal neurons (PyNs), allowing them to exert powerful yet precise control over PyN firing and population output. The importance of proper ChC function is further underscored by the association of ChC connectivity defects with various neurological conditions. Despite this, the cellular mechanisms governing ChC axo-axonic synapse formation remain poorly understood. Here, we identify microglia as key regulators of ChC axonal morphogenesis and AIS synaptogenesis, and show that disease-induced aberrant microglial activation perturbs proper ChC synaptic development/connectivity in the neocortex. In doing so, such findings highlight the therapeutic potential of manipulating microglia to ensure proper brain wiring.


Subject(s)
Axon Initial Segment , GABAergic Neurons , Microglia , Pyramidal Cells , Synapses , Animals , Axon Initial Segment/physiology , GABAergic Neurons/physiology , GABAergic Neurons/ultrastructure , Mice , Microglia/physiology , Pyramidal Cells/physiology , Pyramidal Cells/ultrastructure , Synapses/physiology
7.
Nat Rev Cancer ; 22(2): 102-113, 2022 02.
Article in English | MEDLINE | ID: mdl-34764459

ABSTRACT

Copper is an essential nutrient whose redox properties make it both beneficial and toxic to the cell. Recent progress in studying transition metal signalling has forged new links between researchers of different disciplines that can help translate basic research in the chemistry and biology of copper into clinical therapies and diagnostics to exploit copper-dependent disease vulnerabilities. This concept is particularly relevant in cancer, as tumour growth and metastasis have a heightened requirement for this metal nutrient. Indeed, the traditional view of copper as solely an active site metabolic cofactor has been challenged by emerging evidence that copper is also a dynamic signalling metal and metalloallosteric regulator, such as for copper-dependent phosphodiesterase 3B (PDE3B) in lipolysis, mitogen-activated protein kinase kinase 1 (MEK1) and MEK2 in cell growth and proliferation and the kinases ULK1 and ULK2 in autophagy. In this Perspective, we summarize our current understanding of the connection between copper and cancer and explore how challenges in the field could be addressed by using the framework of cuproplasia, which is defined as regulated copper-dependent cell proliferation and is a representative example of a broad range of metalloplasias. Cuproplasia is linked to a diverse array of cellular processes, including mitochondrial respiration, antioxidant defence, redox signalling, kinase signalling, autophagy and protein quality control. Identifying and characterizing new modes of copper-dependent signalling offers translational opportunities that leverage disease vulnerabilities to this metal nutrient.


Subject(s)
Copper , Neoplasms , Autophagy , Cell Proliferation , Copper/metabolism , Humans , Signal Transduction
8.
Proc Natl Acad Sci U S A ; 118(21)2021 05 25.
Article in English | MEDLINE | ID: mdl-34021083

ABSTRACT

Pancreatic ductal adenocarcinoma (PDAC) is a lethal malignancy with limited treatment options. Although activating mutations of the KRAS GTPase are the predominant dependency present in >90% of PDAC patients, targeting KRAS mutants directly has been challenging in PDAC. Similarly, strategies targeting known KRAS downstream effectors have had limited clinical success due to feedback mechanisms, alternate pathways, and dose-limiting toxicities in normal tissues. Therefore, identifying additional functionally relevant KRAS interactions in PDAC may allow for a better understanding of feedback mechanisms and unveil potential therapeutic targets. Here, we used proximity labeling to identify protein interactors of active KRAS in PDAC cells. We expressed fusions of wild-type (WT) (BirA-KRAS4B), mutant (BirA-KRAS4BG12D), and nontransforming cytosolic double mutant (BirA-KRAS4BG12D/C185S) KRAS with the BirA biotin ligase in murine PDAC cells. Mass spectrometry analysis revealed that RSK1 selectively interacts with membrane-bound KRASG12D, and we demonstrate that this interaction requires NF1 and SPRED2. We find that membrane RSK1 mediates negative feedback on WT RAS signaling and impedes the proliferation of pancreatic cancer cells upon the ablation of mutant KRAS. Our findings link NF1 to the membrane-localized functions of RSK1 and highlight a role for WT RAS signaling in promoting adaptive resistance to mutant KRAS-specific inhibitors in PDAC.


Subject(s)
Carcinoma, Pancreatic Ductal/genetics , Neurofibromin 1/genetics , Proto-Oncogene Proteins p21(ras)/genetics , Ribosomal Protein S6 Kinases, 90-kDa/genetics , Animals , Carcinoma, Pancreatic Ductal/pathology , Cell Line, Tumor , Cell Proliferation/genetics , Humans , Mice , Mutation , Pancreas/pathology , Repressor Proteins/genetics , Signal Transduction/genetics
9.
Curr Opin Neurobiol ; 69: 105-112, 2021 08.
Article in English | MEDLINE | ID: mdl-33862423

ABSTRACT

One of the most intriguing features of inhibitory synapses is the precision by which they innervate their target, not only at the cellular level but also at the subcellular level (i.e. axo-dendritic, axo-somatic, or axo-axonic innervation). In particular, in the cerebellum, cortex, and spinal cord, distinct and highly specialized GABAergic interneurons, such as basket cells, chandelier cells, and GABApre interneurons, form precise axo-axonic synapses, allowing them to directly regulate neuronal output and circuit function. In this article, we summarize our latest knowledge of the cellular and molecular mechanisms that regulate the establishment and maintenance of axo-axonic synapses in these regions of the CNS. We also detail the key roles of the L1CAM family of cell adhesion molecules in such GABAergic subcellular target recognition.


Subject(s)
Axons , Interneurons , Cerebral Cortex , Synapses
10.
Neuron ; 109(10): 1636-1656.e8, 2021 05 19.
Article in English | MEDLINE | ID: mdl-33831348

ABSTRACT

Ample evidence indicates that individuals with intellectual disability (ID) are at increased risk of developing stress-related behavioral problems and mood disorders, yet a mechanistic explanation for such a link remains largely elusive. Here, we focused on characterizing the syndromic ID gene oligophrenin-1 (OPHN1). We find that Ophn1 deficiency in mice markedly enhances helpless/depressive-like behavior in the face of repeated/uncontrollable stress. Strikingly, Ophn1 deletion exclusively in parvalbumin (PV) interneurons in the prelimbic medial prefrontal cortex (PL-mPFC) is sufficient to induce helplessness. This behavioral phenotype is mediated by a diminished excitatory drive onto Ophn1-deficient PL-mPFC PV interneurons, leading to hyperactivity in this region. Importantly, suppressing neuronal activity or RhoA/Rho-kinase signaling in the PL-mPFC reverses helpless behavior. Our results identify OPHN1 as a critical regulator of adaptive behavioral responses to stress and shed light onto the mechanistic links among OPHN1 genetic deficits, mPFC circuit dysfunction, and abnormalities in stress-related behaviors.


Subject(s)
Cytoskeletal Proteins/metabolism , GTPase-Activating Proteins/metabolism , Interneurons/metabolism , Prefrontal Cortex/metabolism , Stress, Psychological/metabolism , Animals , Cytoskeletal Proteins/deficiency , Cytoskeletal Proteins/genetics , GTPase-Activating Proteins/deficiency , GTPase-Activating Proteins/genetics , HEK293 Cells , Helplessness, Learned , Humans , Interneurons/physiology , Mice , Mice, Inbred C57BL , Parvalbumins/genetics , Parvalbumins/metabolism , Prefrontal Cortex/cytology , Prefrontal Cortex/physiology , Stress, Psychological/physiopathology , Synaptic Transmission
11.
J Exp Med ; 217(10)2020 10 05.
Article in English | MEDLINE | ID: mdl-32667673

ABSTRACT

C-C chemokine receptor type 2 (CCR2) is expressed on monocytes and facilitates their recruitment to tumors. Though breast cancer cells also express CCR2, its functions in these cells are unclear. We found that Ccr2 deletion in cancer cells led to reduced tumor growth and approximately twofold longer survival in an orthotopic, isograft breast cancer mouse model. Deletion of Ccr2 in cancer cells resulted in multiple alterations associated with better immune control: increased infiltration and activation of cytotoxic T lymphocytes (CTLs) and CD103+ cross-presenting dendritic cells (DCs), as well as up-regulation of MHC class I and down-regulation of checkpoint regulator PD-L1 on the cancer cells. Pharmacological or genetic targeting of CCR2 increased cancer cell sensitivity to CTLs and enabled the cancer cells to induce DC maturation toward the CD103+ subtype. Consistently, Ccr2-/- cancer cells did not induce immune suppression in Batf3-/- mice lacking CD103+ DCs. Our results establish that CCR2 signaling in cancer cells can orchestrate suppression of the immune response.


Subject(s)
Adaptive Immunity/immunology , Immune Tolerance , Mammary Neoplasms, Experimental/immunology , Receptors, CCR2/physiology , Adaptive Immunity/physiology , Animals , Apoptosis , B7-H1 Antigen/metabolism , Dendritic Cells/immunology , Dendritic Cells/physiology , Female , Histocompatibility Antigens Class I/metabolism , Immune Tolerance/immunology , Immune Tolerance/physiology , Interferons/metabolism , Mice , Mice, Inbred C57BL , Receptors, CCR2/immunology , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Cytotoxic/physiology
12.
Trends Neurosci ; 43(8): 565-580, 2020 08.
Article in English | MEDLINE | ID: mdl-32564887

ABSTRACT

Chandelier cells (ChCs) are a unique type of GABAergic interneuron that selectively innervate the axon initial segment (AIS) of excitatory pyramidal neurons; the subcellular domain where action potentials are initiated. The proper genesis and maturation of ChCs is critical for regulating neural ensemble firing in the neocortex throughout development and adulthood. Recently, genetic and molecular studies have shed new light on the complex innerworkings of ChCs in health and disease. This review presents an overview of recent studies on the developmental origins, migratory properties, and morphology of ChCs. In addition, attention is given to newly identified molecules regulating ChC morphogenesis and connectivity as well as recent work linking ChC dysfunction to neural disorders, including schizophrenia, epilepsy, and autism spectrum disorder (ASD).


Subject(s)
Autism Spectrum Disorder , Neocortex , Adult , Humans , Interneurons , Neurons , Pyramidal Cells
13.
Cancer Cell ; 36(5): 468-470, 2019 Nov 11.
Article in English | MEDLINE | ID: mdl-31715130

ABSTRACT

Stress has long been suspected to negatively influence cancer mortality, yet the molecular mechanisms responsible for this effect have only recently been identified. A new study identifies a stress-induced response in dendritic cells-the activation of the glucocorticoid-inducible transcriptional regulator TSC22D3-as a potent, immunosuppressive effect of stress on cancer.


Subject(s)
Dendritic Cells/immunology , Glucocorticoids/metabolism , Immunotherapy , Neoplasms/therapy , Stress, Psychological/immunology , Dendritic Cells/metabolism , Disease Progression , Glucocorticoids/immunology , Humans , Immune Tolerance , Neoplasms/immunology , Neoplasms/mortality , Neoplasms/psychology , Stress, Psychological/psychology , Transcription Factors/immunology , Transcription Factors/metabolism , Treatment Outcome
14.
Neuron ; 102(2): 358-372.e9, 2019 04 17.
Article in English | MEDLINE | ID: mdl-30846310

ABSTRACT

Among the diverse interneuron subtypes in the neocortex, chandelier cells (ChCs) are the only population that selectively innervate pyramidal neurons (PyNs) at their axon initial segment (AIS), the site of action potential initiation, allowing them to exert powerful control over PyN output. Yet, mechanisms underlying their subcellular innervation of PyN AISs are unknown. To identify molecular determinants of ChC/PyN AIS innervation, we performed an in vivo RNAi screen of PyN-expressed axonal cell adhesion molecules (CAMs) and select Ephs/ephrins. Strikingly, we found the L1 family member L1CAM to be the only molecule required for ChC/PyN AIS innervation. Further, we show that L1CAM is required during both the establishment and maintenance of innervation, and that selective innervation of PyN AISs by ChCs requires AIS anchoring of L1CAM by the cytoskeletal ankyrin-G/ßIV-spectrin complex. Thus, our findings identify PyN-expressed L1CAM as a critical CAM required for innervation of neocortical PyN AISs by ChCs. VIDEO ABSTRACT.


Subject(s)
Axons/metabolism , GABAergic Neurons/metabolism , Interneurons/metabolism , Neural Cell Adhesion Molecule L1/metabolism , Pyramidal Cells/metabolism , Animals , Ankyrins/metabolism , Cell Adhesion , Cell Adhesion Molecules/metabolism , Ephrins/metabolism , Mice , Neocortex/cytology , Neocortex/metabolism , Nerve Tissue Proteins/metabolism , Receptor, EphA1/metabolism , Spectrin/metabolism , Synapses
15.
J Cell Biol ; 216(12): 4313-4330, 2017 12 04.
Article in English | MEDLINE | ID: mdl-29089377

ABSTRACT

Throughout life, stem cells in the ventricular-subventricular zone generate neuroblasts that migrate via the rostral migratory stream (RMS) to the olfactory bulb, where they differentiate into local interneurons. Although progress has been made toward identifying extracellular factors that guide the migration of these cells, little is known about the intracellular mechanisms that govern the dynamic reshaping of the neuroblasts' morphology required for their migration along the RMS. In this study, we identify DOCK7, a member of the DOCK180-family, as a molecule essential for tangential neuroblast migration in the postnatal mouse forebrain. DOCK7 regulates the migration of these cells by controlling both leading process (LP) extension and somal translocation via distinct pathways. It controls LP stability/growth via a Rac-dependent pathway, likely by modulating microtubule networks while also regulating F-actin remodeling at the cell rear to promote somal translocation via a previously unrecognized myosin phosphatase-RhoA-interacting protein-dependent pathway. The coordinated action of both pathways is required to ensure efficient neuroblast migration along the RMS.


Subject(s)
Guanine Nucleotide Exchange Factors/genetics , Myosin-Light-Chain Phosphatase/genetics , Neurons/metabolism , Prosencephalon/metabolism , Proto-Oncogene Proteins c-akt/genetics , rho GTP-Binding Proteins/genetics , Actins/genetics , Actins/metabolism , Animals , Animals, Newborn , Cell Differentiation , Cell Line, Tumor , Cell Movement , Embryo, Mammalian , GTPase-Activating Proteins , Gene Expression Regulation, Developmental , Guanine Nucleotide Exchange Factors/metabolism , HEK293 Cells , HeLa Cells , Humans , Mice , Microtubules/metabolism , Microtubules/ultrastructure , Myosin-Light-Chain Phosphatase/metabolism , Neurons/ultrastructure , Primary Cell Culture , Prosencephalon/cytology , Prosencephalon/growth & development , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction , rho GTP-Binding Proteins/metabolism , rhoA GTP-Binding Protein
16.
J Clin Invest ; 125(8): 3163-77, 2015 Aug 03.
Article in English | MEDLINE | ID: mdl-26214522

ABSTRACT

The X-linked neurological disorder Rett syndrome (RTT) presents with autistic features and is caused primarily by mutations in a transcriptional regulator, methyl CpG-binding protein 2 (MECP2). Current treatment options for RTT are limited to alleviating some neurological symptoms; hence, more effective therapeutic strategies are needed. We identified the protein tyrosine phosphatase PTP1B as a therapeutic candidate for treatment of RTT. We demonstrated that the PTPN1 gene, which encodes PTP1B, was a target of MECP2 and that disruption of MECP2 function was associated with increased levels of PTP1B in RTT models. Pharmacological inhibition of PTP1B ameliorated the effects of MECP2 disruption in mouse models of RTT, including improved survival in young male (Mecp2-/y) mice and improved behavior in female heterozygous (Mecp2-/+) mice. We demonstrated that PTP1B was a negative regulator of tyrosine phosphorylation of the tyrosine kinase TRKB, the receptor for brain-derived neurotrophic factor (BDNF). Therefore, the elevated PTP1B that accompanies disruption of MECP2 function in RTT represents a barrier to BDNF signaling. Inhibition of PTP1B led to increased tyrosine phosphorylation of TRKB in the brain, which would augment BDNF signaling. This study presents PTP1B as a mechanism-based therapeutic target for RTT, validating a unique strategy for treating the disease by modifying signal transduction pathways with small-molecule drugs.


Subject(s)
Enzyme Inhibitors/pharmacology , Protein Tyrosine Phosphatase, Non-Receptor Type 1/antagonists & inhibitors , Protein Tyrosine Phosphatase, Non-Receptor Type 1/metabolism , Rett Syndrome/drug therapy , Signal Transduction/drug effects , Animals , Brain-Derived Neurotrophic Factor/genetics , Brain-Derived Neurotrophic Factor/metabolism , Disease Models, Animal , Female , Male , Methyl-CpG-Binding Protein 2/genetics , Methyl-CpG-Binding Protein 2/metabolism , Mice , Mice, Inbred CBA , Mice, Mutant Strains , Phosphorylation/drug effects , Phosphorylation/genetics , Protein Tyrosine Phosphatase, Non-Receptor Type 1/genetics , Receptor, trkB/genetics , Receptor, trkB/metabolism , Rett Syndrome/enzymology , Rett Syndrome/genetics , Rett Syndrome/pathology , Signal Transduction/genetics
17.
Genes Dev ; 29(3): 250-61, 2015 Feb 01.
Article in English | MEDLINE | ID: mdl-25644601

ABSTRACT

The mechanisms by which TGF-ß promotes lung adenocarcinoma (ADC) metastasis are largely unknown. Here, we report that in lung ADC cells, TGF-ß potently induces expression of DOCK4, but not other DOCK family members, via the Smad pathway and that DOCK4 induction mediates TGF-ß's prometastatic effects by enhancing tumor cell extravasation. TGF-ß-induced DOCK4 stimulates lung ADC cell protrusion, motility, and invasion without affecting epithelial-to-mesenchymal transition. These processes, which are fundamental to tumor cell extravasation, are driven by DOCK4-mediated Rac1 activation, unveiling a novel link between TGF-ß and Rac1. Thus, our findings uncover the atypical Rac1 activator DOCK4 as a key component of the TGF-ß/Smad pathway that promotes lung ADC cell extravasation and metastasis.


Subject(s)
Adenocarcinoma/physiopathology , GTPase-Activating Proteins/metabolism , Lung Neoplasms/physiopathology , Signal Transduction , Smad Proteins/metabolism , Transforming Growth Factor beta/metabolism , Adenocarcinoma of Lung , Animals , Cell Line, Tumor , GTPase-Activating Proteins/genetics , Gene Expression Regulation, Neoplastic , Humans , Mice , Neoplasm Metastasis
18.
Nature ; 519(7544): 455-9, 2015 Mar 26.
Article in English | MEDLINE | ID: mdl-25600269

ABSTRACT

Appropriate responses to an imminent threat brace us for adversities. The ability to sense and predict threatening or stressful events is essential for such adaptive behaviour. In the mammalian brain, one putative stress sensor is the paraventricular nucleus of the thalamus (PVT), an area that is readily activated by both physical and psychological stressors. However, the role of the PVT in the establishment of adaptive behavioural responses remains unclear. Here we show in mice that the PVT regulates fear processing in the lateral division of the central amygdala (CeL), a structure that orchestrates fear learning and expression. Selective inactivation of CeL-projecting PVT neurons prevented fear conditioning, an effect that can be accounted for by an impairment in fear-conditioning-induced synaptic potentiation onto somatostatin-expressing (SOM(+)) CeL neurons, which has previously been shown to store fear memory. Consistently, we found that PVT neurons preferentially innervate SOM(+) neurons in the CeL, and stimulation of PVT afferents facilitated SOM(+) neuron activity and promoted intra-CeL inhibition, two processes that are critical for fear learning and expression. Notably, PVT modulation of SOM(+) CeL neurons was mediated by activation of the brain-derived neurotrophic factor (BDNF) receptor tropomysin-related kinase B (TrkB). As a result, selective deletion of either Bdnf in the PVT or Trkb in SOM(+) CeL neurons impaired fear conditioning, while infusion of BDNF into the CeL enhanced fear learning and elicited unconditioned fear responses. Our results demonstrate that the PVT-CeL pathway constitutes a novel circuit essential for both the establishment of fear memory and the expression of fear responses, and uncover mechanisms linking stress detection in PVT with the emergence of adaptive behaviour.


Subject(s)
Central Amygdaloid Nucleus/physiology , Fear/physiology , Neural Pathways/physiology , Thalamus/physiology , Animals , Brain-Derived Neurotrophic Factor/metabolism , Central Amygdaloid Nucleus/cytology , Conditioning, Psychological/physiology , Fear/psychology , Female , Male , Memory/physiology , Mice , Neural Pathways/cytology , Neuronal Plasticity , Neurons/metabolism , Receptor, trkB/metabolism , Somatostatin/metabolism , Thalamus/cytology , Time Factors
19.
J Neurosci ; 34(26): 8665-71, 2014 Jun 25.
Article in English | MEDLINE | ID: mdl-24966368

ABSTRACT

At glutamatergic synapses, local endocytic recycling of AMPA receptors (AMPARs) is important for the supply of a mobile pool of AMPARs required for synaptic potentiation. This local recycling of AMPARs critically relies on the presence of an endocytic zone (EZ) near the postsynaptic density (PSD). The precise mechanisms that couple the EZ to the PSD still remain largely elusive, with the large GTPase Dynamin-3 and the multimeric PSD adaptor protein Homer1 as the two main players identified. Here, we demonstrate that a physical interaction between the X-linked mental retardation protein oligophrenin-1 (OPHN1) and Homer1b/c is crucial for the positioning of the EZ adjacent to the PSD, and present evidence that this interaction is important for OPHN1's role in controlling activity-dependent strengthening of excitatory synapses in the rat hippocampus. Disruption of the OPHN1-Homer1b/c interaction causes a displacement of EZs from the PSD, along with impaired AMPAR recycling and reduced AMPAR accumulation at synapses, in both basal conditions and conditions that can induce synaptic potentiation. Together, our findings unveil a novel role for OPHN1 as an interaction partner of Homer1b/c in spine EZ positioning, and provide new mechanistic insight into how genetic deficits in OPHN1 can lead to impaired synapse maturation and plasticity.


Subject(s)
Carrier Proteins/metabolism , Cytoskeletal Proteins/metabolism , Endocytosis/physiology , GTPase-Activating Proteins/metabolism , Neuronal Plasticity/physiology , Nuclear Proteins/metabolism , Synapses/metabolism , Animals , Dendritic Spines/metabolism , Hippocampus/metabolism , Homer Scaffolding Proteins , Neurons/metabolism , Rats , Receptors, AMPA/metabolism , Synaptic Transmission/physiology
20.
Cell Rep ; 6(2): 254-63, 2014 Jan 30.
Article in English | MEDLINE | ID: mdl-24440718

ABSTRACT

Chandelier cells (ChCs), typified by their unique axonal morphology, are the most distinct interneurons present in cortical circuits. Via their distinctive axonal terminals, called cartridges, these cells selectively target the axon initial segment of pyramidal cells and control action potential initiation; however, the mechanisms that govern the characteristic ChC axonal structure have remained elusive. Here, by employing an in utero electroporation-based method that enables genetic labeling and manipulation of ChCs in vivo, we identify DOCK7, a member of the DOCK180 family, as a molecule essential for ChC cartridge and bouton development. Furthermore, we present evidence that DOCK7 functions as a cytoplasmic activator of the schizophrenia-associated ErbB4 receptor tyrosine kinase and that DOCK7 modulates ErbB4 activity to control ChC cartridge and bouton development. Thus, our findings define DOCK7 and ErbB4 as key components of a pathway that controls the morphological differentiation of ChCs, with implications for the pathogenesis of schizophrenia.


Subject(s)
Cerebral Cortex/growth & development , Guanine Nucleotide Exchange Factors/metabolism , Interneurons/metabolism , Presynaptic Terminals/metabolism , Animals , Cerebral Cortex/cytology , Cerebral Cortex/metabolism , ErbB Receptors/genetics , ErbB Receptors/metabolism , GTPase-Activating Proteins , Guanine Nucleotide Exchange Factors/genetics , Mice , Receptor, ErbB-4
SELECTION OF CITATIONS
SEARCH DETAIL
...