Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 93
Filter
Add more filters










Publication year range
1.
Front Mol Neurosci ; 16: 1305574, 2023.
Article in English | MEDLINE | ID: mdl-38106879

ABSTRACT

Despite structural similarity with other tumor necrosis factor receptor superfamily (TNFRSF) members, the p75 neurotrophin receptor (p75NTR, TNFR16) mediates pleiotropic biological functions not shared with other TNFRs. The high level of p75NTR expression in the nervous system instead of immune cells, its utilization of co-receptors, and its interaction with soluble dimeric, rather than soluble or cell-tethered trimeric ligands are all characteristics which distinguish it from most other TNFRs. Here, we compare these attributes to other members of the TNFR superfamily. In addition, we describe the recent evolutionary adaptation in B7-1 (CD80), an immunoglobulin (Ig) superfamily member, which allows engagement to neuronally-expressed p75NTR. B7-1-mediated binding to p75NTR occurs in humans and other primates, but not lower mammals due to specific sequence changes that evolved recently in primate B7-1. This discovery highlights an additional mechanism by which p75NTR can respond to inflammatory cues and trigger synaptic elimination in the brain through engagement of B7-1, which was considered to be immune-restricted. These observations suggest p75NTR does share commonality with other immune co-modulatory TNFR family members, by responding to immunoregulatory cues. The evolution of primate B7-1 to bind and elicit p75NTR-mediated effects on neuronal morphology and function are discussed in relationship to immune-driven modulation of synaptic actions during injury or inflammation.

2.
J Clin Invest ; 132(22)2022 11 15.
Article in English | MEDLINE | ID: mdl-36107635

ABSTRACT

Cell surface receptors, ligands, and adhesion molecules underlie development, circuit formation, and synaptic function of the central nervous system and represent important therapeutic targets for many neuropathologies. The functional contributions of interactions between cell surface proteins of neurons and nonneuronal cells have not been fully addressed. Using an unbiased protein-protein interaction screen, we showed that the human immunomodulatory ligand B7-1 (hB7-1) interacts with the p75 neurotrophin receptor (p75NTR) and that the B7-1:p75NTR interaction is a recent evolutionary adaptation present in humans and other primates, but absent in mice, rats, and other lower mammals. The surface of hB7-1 that engages p75NTR overlaps with the hB7-1 surface involved in CTLA-4/CD28 recognition, and these molecules directly compete for binding to p75NTR. Soluble or membrane-bound hB7-1 altered dendritic morphology of cultured hippocampal neurons, with loss of the postsynaptic protein PSD95 in a p75NTR-dependent manner. Abatacept, an FDA-approved therapeutic (CTLA-4-hFc fusion) inhibited these processes. In vivo injection of hB7-1 into the murine subiculum, a hippocampal region affected in Alzheimer's disease, resulted in p75NTR-dependent pruning of dendritic spines. Here, we report the biochemical interaction between B7-1 and p75NTR, describe biological effects on neuronal morphology, and identify a therapeutic opportunity for treatment of neuroinflammatory diseases.


Subject(s)
B7-1 Antigen , Neurons , Receptor, Nerve Growth Factor , Receptors, Nerve Growth Factor , Synapses , Animals , Humans , Mice , Rats , CTLA-4 Antigen/metabolism , Neurons/metabolism , Receptor, Nerve Growth Factor/genetics , Receptor, Nerve Growth Factor/metabolism , Receptors, Nerve Growth Factor/genetics , Receptors, Nerve Growth Factor/metabolism , B7-1 Antigen/metabolism , Synapses/metabolism
3.
Mol Psychiatry ; 26(3): 927-940, 2021 03.
Article in English | MEDLINE | ID: mdl-31988435

ABSTRACT

Social memory processing requires functional CA2 neurons, however the specific mechanisms that regulate their activity are poorly understood. Here, we document that SorCS2, a member of the family of the Vps10 family of sorting receptors, is highly expressed in pyramidal neurons of CA2, as well as ventral CA1, a circuit implicated in social memory. SorCS2 specifically localizes to the postsynaptic density and endosomes within dendritic spines of CA2 neurons. We have discovered that SorCS2 is a selective regulator of NMDA receptor surface trafficking in hippocampal neurons, without altering AMPA receptor trafficking. In addition, SorCS2 regulates dendritic spine density in CA2 neurons where SorCS2 expression is enriched, but not in dorsal CA1 neurons, which normally express very low levels of this protein. To specifically test the role of SorCS2 in behavior, we generated a novel SorCS2-deficient mouse, and identify a significant social memory deficit, with no change in sociability, olfaction, anxiety, or several hippocampal-dependent behaviors. Mutations in sorCS2 have been associated with bipolar disease, schizophrenia, and attention deficient-hyperactivity disorder, and abnormalities in social memory are core components of these neuropsychiatric conditions. Thus, our findings provide a new mechanism for social memory formation, through regulating synaptic receptor trafficking in pyramidal neurons by SorCS2.


Subject(s)
Memory , Nerve Tissue Proteins , Pyramidal Cells , Receptors, Cell Surface , Receptors, N-Methyl-D-Aspartate , Animals , Dendritic Spines/metabolism , Hippocampus/metabolism , Mice , Neurons/metabolism , Pyramidal Cells/metabolism , Receptors, N-Methyl-D-Aspartate/genetics , Receptors, N-Methyl-D-Aspartate/metabolism
4.
Metallomics ; 12(8): 1208-1219, 2020 08 19.
Article in English | MEDLINE | ID: mdl-32744273

ABSTRACT

Human brain derived neurotrophic factor (BDNF) encodes a protein product consisting of a C-terminal mature domain (mature BDNF) and an N-terminal prodomain, which is an intrinsically disordered protein. A common single nucleotide polymorphism in humans results in a methionine substitution for valine at position 66 of the prodomain, and is associated with memory deficits, depression and anxiety disorders. The BDNF Met66 prodomain, but not the Val66 prodomain, promotes rapid structural remodeling of hippocampal neurons' growth cones and dendritic spines by interacting directly with the SorCS2 receptor. While it has been reported that the Met66 and Val66 prodomains exhibit only modest differences in structural propensities in the apo state, here we show that Val66 and Met66 prodomains differentially bind zinc (Zn). Zn2+ binds with higher affinity and more broadly impacts residues on the Met66 prodomain compared to the Val66 prodomain as shown by NMR and ITC. Zn2+ binding to the Met66 and Val66 prodomains results in distinct conformational and macroscopic differences observed by NMR, light scattering and cryoEM. To determine if Zn2+ mediated conformational change in the Met66 prodomain is required for biological effect, we mutated His40, a Zn2+ binding site, and observed a loss of Met66 prodomain bioactivity. As the His40 site is distant from the known region of the prodomain involved in receptor binding, we suggest that Met66 prodomain bioactivity involves His40 mediated stabilization of the multimeric structure. Our results point to the necessity of a Zn2+-mediated higher order molecular assembly of the Met66 prodomain to mediate neuronal remodeling.


Subject(s)
Brain-Derived Neurotrophic Factor/chemistry , Zinc/chemistry , Binding Sites , Magnetic Resonance Spectroscopy , Nerve Tissue Proteins/chemistry , Protein Binding
5.
Front Physiol ; 10: 455, 2019.
Article in English | MEDLINE | ID: mdl-31105581

ABSTRACT

The actions of BDNF (Brain-derived Neurotrophic Factor) in regulating neuronal development and modulating synaptic activity have been extensively studied and well established. Equally important roles for this growth factor have been uncovered in the cardiovascular system, through the examination of gene targeted animals to define critical actions in development, and to the unexpected roles of BDNF in modulating the response of the heart and vasculature to injury. Here we review the compartmentally distinct realm of cardiac myocytes, vascular smooth muscle cells, endothelial cells, and hematopoietic cells, focusing upon the actions of BDNF to modulate contractility, migration, neoangiogenesis, apoptosis and survival. These studies indicate that BDNF is an important growth factor which directs the response of the cardiovascular system to acute and chronic injury.

6.
Trends Neurosci ; 41(11): 765-767, 2018 11.
Article in English | MEDLINE | ID: mdl-30219601

ABSTRACT

In 1996, Hyejin Kang and Erin Schuman, in search of new functions for the secreted growth factor brain-derived neurotrophic factor (BDNF), identified the protein synthesis requirement of BDNF in regulating synaptic plasticity. This landmark paper identified one of the first tractable pathways in the quest to dissect the complex process of synaptic remodeling and revealed the critical role for this neurotrophin in regulating long-term memory.


Subject(s)
Brain-Derived Neurotrophic Factor/metabolism , Neuronal Plasticity/physiology , Synapses/metabolism , Synaptic Transmission/physiology , Animals , Long-Term Potentiation/physiology , Long-Term Synaptic Depression/physiology , Protein Biosynthesis/physiology , Rats
8.
Neuron ; 99(1): 163-178.e6, 2018 07 11.
Article in English | MEDLINE | ID: mdl-29909994

ABSTRACT

A human variant in the BDNF gene (Val66Met; rs6265) is associated with impaired fear extinction. Using super-resolution imaging, we demonstrate that the BDNF Met prodomain disassembles dendritic spines and eliminates synapses in hippocampal neurons. In vivo, ventral CA1 (vCA1) hippocampal neurons undergo similar morphological changes dependent on their transient co-expression of a SorCS2/p75NTR receptor complex during peri-adolescence. BDNF Met prodomain infusion into the vCA1 during this developmental time frame reduces dendritic spine density and prelimbic (PL) projections, impairing cued fear extinction. Adolescent BdnfMet/Met mice display similar spine and PL innervation deficits. Using fiber photometry, we found that, in wild-type mice, vCA1 neurons projecting to the PL encode extinction by enhancing neural activity in threat anticipation and rapidly subsiding their response. This adaptation is absent in BDNFMet/Met mice. We conclude that the BDNF Met prodomain renders vCA1-PL projection neurons underdeveloped, preventing their capacity for subsequent circuit modulation necessary for fear extinction. VIDEO ABSTRACT.


Subject(s)
Brain-Derived Neurotrophic Factor/genetics , CA1 Region, Hippocampal/pathology , Dendritic Spines/pathology , Extinction, Psychological , Fear , Neurons/pathology , Synapses/pathology , Animals , CA1 Region, Hippocampal/physiopathology , Mice , Polymorphism, Single Nucleotide
9.
eNeuro ; 5(1)2018.
Article in English | MEDLINE | ID: mdl-29349290

ABSTRACT

Neurotrophins play critical roles in the survival, maintenance and death of neurons. In particular, proneurotrophins have been shown to mediate cell death following brain injury induced by status epilepticus (SE) in rats. Previous studies have shown that pilocarpine-induced seizures lead to increased levels of proNGF, which binds to the p75NTR-sortilin receptor complex to elicit apoptosis. A screen to identify compounds that block proNGF binding and uptake into cells expressing p75 and sortilin identified lithium citrate as a potential inhibitor of proNGF and p75NTR-mediated cell death. In this study, we demonstrate that low, submicromolar doses of lithium citrate effectively inhibited proNGF-induced cell death in cultured neurons and protected hippocampal neurons following pilocarpine-induced SE in vivo. We analyzed specific mechanisms by which lithium citrate afforded neuroprotection and determined that lithium citrate prevented the association and internalization of the p75NTR-sortilin receptor complex. Our results demonstrate a novel mechanism by which low-dose treatments of lithium citrate are effective in attenuating p75NTR-mediated cell death in vitro and in vivo.


Subject(s)
Adaptor Proteins, Vesicular Transport/metabolism , Cell Death/drug effects , Citrates/pharmacology , Neurons/drug effects , Neuroprotective Agents/pharmacology , Receptors, Nerve Growth Factor/metabolism , Animals , Cell Death/physiology , Cells, Cultured , Dose-Response Relationship, Drug , Hippocampus/drug effects , Hippocampus/metabolism , Hippocampus/pathology , Humans , Male , Neurons/metabolism , Neurons/pathology , Pilocarpine , Rats , Sf9 Cells , Status Epilepticus/drug therapy , Status Epilepticus/metabolism , Status Epilepticus/pathology
10.
Neuropsychopharmacology ; 43(5): 1088-1098, 2018 04.
Article in English | MEDLINE | ID: mdl-29052614

ABSTRACT

Growing recognition of persistent cognitive defects associated with electroconvulsive therapy (ECT), a highly effective and commonly used antidepressant treatment, has spurred interest in identifying its mechanism of action to guide development of safer treatment options. However, as repeated seizure activity elicits a bewildering array of electrophysiological and biochemical effects, this goal has remained elusive. We have examined whether deletion of Narp, an immediate early gene induced by electroconvulsive seizures (ECS), blocks its antidepressant efficacy. Based on multiple measures, we infer that Narp knockout mice undergo normal seizure activity in this paradigm, yet fail to display antidepressant-like behavioral effects of ECS. Although Narp deletion does not suppress ECS-induced proliferation in the dentate gyrus, it blocks dendritic outgrowth of immature granule cell neurons in the dentate molecular layer induced by ECS. Taken together, these findings indicate that Narp contributes to the antidepressant action of ECT and implicate the ability of ECS to induce dendritic arborization of differentiating granule cells as a relevant step in eliciting this response.


Subject(s)
C-Reactive Protein/physiology , Cell Proliferation/physiology , Electroshock , Nerve Tissue Proteins/physiology , Neuronal Plasticity/physiology , Seizures/physiopathology , Animals , C-Reactive Protein/genetics , Dentate Gyrus/physiology , Male , Mice , Mice, Knockout , Nerve Tissue Proteins/genetics , Neurons/physiology
11.
Am J Psychiatry ; 174(12): 1203-1213, 2017 12 01.
Article in English | MEDLINE | ID: mdl-29084453

ABSTRACT

OBJECTIVE: Adolescence is a developmental stage in which the incidence of psychiatric disorders, such as anxiety disorders, peaks. Selective serotonin reuptake inhibitors (SSRIs) are the main class of agents used to treat anxiety disorders. However, the impact of SSRIs on the developing brain during adolescence remains unknown. The authors assessed the impact of developmentally timed SSRI administration in a genetic mouse model displaying elevated anxiety-like behaviors. METHOD: Knock-in mice containing a common human single-nucleotide polymorphism (Val66Met; rs6265) in brain-derived neurotrophic factor (BDNF), a growth factor implicated in the mechanism of action of SSRIs, were studied based on their established phenotype of increased anxiety-like behavior. Timed administration of fluoxetine was delivered during one of three developmental periods (postnatal days 21-42, 40-61, or 60-81), spanning the transition from childhood to adulthood. Neurochemical and anxiety-like behavioral analyses were performed. RESULTS: We identified a "sensitive period" during periadolescence (postnatal days 21-42) in which developmentally timed fluoxetine administration rescued anxiety-like phenotypes in BDNF Val66Met mice in adulthood. Compared with littermate controls, BDNFMet/Met mice exhibited diminished maturation of serotonergic fibers projecting particularly to the prefrontal cortex, as well as decreased expression of the serotonergic trophic factor S100B in the dorsal raphe. Interestingly, deficient serotonergic innervation, as well as S100B levels, were rescued with fluoxetine administration during periadolescence. CONCLUSIONS: These findings suggest that SSRI administration during a "sensitive period" during periadolescence leads to long-lasting anxiolytic effects in a genetic mouse model of elevated anxiety-like behaviors. These persistent effects highlight the role of BDNF in the maturation of the serotonin system and the capacity to enhance its development through a pharmacological intervention.


Subject(s)
Anti-Anxiety Agents/pharmacology , Anti-Anxiety Agents/therapeutic use , Anxiety/drug therapy , Anxiety/genetics , Brain-Derived Neurotrophic Factor/genetics , Fluoxetine/pharmacology , Fluoxetine/therapeutic use , Age Factors , Animals , Dorsal Raphe Nucleus/drug effects , Dorsal Raphe Nucleus/metabolism , Eating/drug effects , Fear/drug effects , Gene Knock-In Techniques , Male , Maze Learning/drug effects , Mice , Polymorphism, Single Nucleotide/genetics , Prefrontal Cortex/drug effects , S100 Calcium Binding Protein beta Subunit/metabolism , Serotonergic Neurons/drug effects , Selective Serotonin Reuptake Inhibitors/pharmacology , Selective Serotonin Reuptake Inhibitors/therapeutic use
12.
JCI Insight ; 2(9)2017 May 04.
Article in English | MEDLINE | ID: mdl-28469074

ABSTRACT

Motor dysfunction is a prominent and disabling feature of Huntington's disease (HD), but the molecular mechanisms that dictate its onset and progression are unknown. The N-methyl-D-aspartate receptor 2A (NR2A) subunit regulates motor skill development and synaptic plasticity in medium spiny neurons (MSNs) of the striatum, cells that are most severely impacted by HD. Here, we document reduced NR2A receptor subunits on the dendritic membranes and at the synapses of MSNs in zQ175 mice that model HD. We identify that SorCS2, a vacuolar protein sorting 10 protein-domain (VPS10P-domain) receptor, interacts with VPS35, a core component of retromer, thereby regulating surface trafficking of NR2A in MSNs. In the zQ175 striatum, SorCS2 is markedly decreased in an age- and allele-dependent manner. Notably, SorCS2 selectively interacts with mutant huntingtin (mtHTT), but not WT huntingtin (wtHTT), and is mislocalized to perinuclear clusters in striatal neurons of human HD patients and zQ175 mice. Genetic deficiency of SorCS2 accelerates the onset and exacerbates the motor coordination deficit of zQ175 mice. Together, our results identify SorCS2 as an interacting protein of mtHTT and demonstrate that impaired SorCS2-mediated NR2A subunit trafficking to dendritic surface of MSNs is, to our knowledge, a novel mechanism contributing to motor coordination deficits of HD.

13.
Nature ; 538(7623): 99-103, 2016 Oct 06.
Article in English | MEDLINE | ID: mdl-27680698

ABSTRACT

Brain-derived neurotrophic factor (BDNF) and its receptor TrkB are crucial for many forms of neuronal plasticity, including structural long-term potentiation (sLTP), which is a correlate of an animal's learning. However, it is unknown whether BDNF release and TrkB activation occur during sLTP, and if so, when and where. Here, using a fluorescence resonance energy transfer-based sensor for TrkB and two-photon fluorescence lifetime imaging microscopy, we monitor TrkB activity in single dendritic spines of CA1 pyramidal neurons in cultured murine hippocampal slices. In response to sLTP induction, we find fast (onset < 1 min) and sustained (>20 min) activation of TrkB in the stimulated spine that depends on NMDAR (N-methyl-d-aspartate receptor) and CaMKII signalling and on postsynaptically synthesized BDNF. We confirm the presence of postsynaptic BDNF using electron microscopy to localize endogenous BDNF to dendrites and spines of hippocampal CA1 pyramidal neurons. Consistent with these findings, we also show rapid, glutamate-uncaging-evoked, time-locked BDNF release from single dendritic spines using BDNF fused to superecliptic pHluorin. We demonstrate that this postsynaptic BDNF-TrkB signalling pathway is necessary for both structural and functional LTP. Together, these findings reveal a spine-autonomous, autocrine signalling mechanism involving NMDAR-CaMKII-dependent BDNF release from stimulated dendritic spines and subsequent TrkB activation on these same spines that is crucial for structural and functional plasticity.


Subject(s)
Autocrine Communication , Brain-Derived Neurotrophic Factor/metabolism , Dendritic Spines/metabolism , Membrane Glycoproteins/metabolism , Protein-Tyrosine Kinases/metabolism , Signal Transduction , Animals , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Dendritic Spines/ultrastructure , Enzyme Activation , Female , Fluorescence Resonance Energy Transfer , Glutamic Acid/metabolism , Green Fluorescent Proteins , HeLa Cells , Hippocampus/cytology , Hippocampus/metabolism , Hippocampus/ultrastructure , Humans , Long-Term Potentiation , Male , Mice , Mice, Inbred C57BL , Microscopy, Electron , Microscopy, Fluorescence, Multiphoton , Post-Synaptic Density/metabolism , Pyramidal Cells/metabolism , Pyramidal Cells/ultrastructure , Rats , Receptors, N-Methyl-D-Aspartate/metabolism , Tissue Culture Techniques
14.
eNeuro ; 3(1)2016.
Article in English | MEDLINE | ID: mdl-27057559

ABSTRACT

Brain-derived neurotrophic factor (BDNF) levels are elevated after status epilepticus (SE), leading to activation of multiple signaling pathways, including the janus kinase/signal transducer and activator of transcription pathway that mediates a decrease in GABAA receptor α1 subunits in the hippocampus (Lund et al., 2008). While BDNF can signal via its pro or mature form, the relative contribution of these forms to signaling after SE is not fully known. In the current study, we investigate changes in proBDNF levels acutely after SE in C57BL/6J mice. In contrast to previous reports (Unsain et al., 2008; Volosin et al., 2008; VonDran et al., 2014), our studies found that levels of proBDNF in the hippocampus are markedly elevated as early as 3 h after SE onset and remain elevated for 7 d. Immunohistochemistry studies indicate that seizure-induced BDNF localizes to all hippocampal subfields, predominantly in principal neurons and also in astrocytes. Analysis of the proteolytic machinery that cleaves proBDNF to produce mature BDNF demonstrates that acutely after SE there is a decrease in tissue plasminogen activator and an increase in plasminogen activator inhibitor-1 (PAI-1), an inhibitor of extracellular and intracellular cleavage, which normalizes over the first week after SE. In vitro treatment of hippocampal slices from animals 24 h after SE with a PAI-1 inhibitor reduces proBDNF levels. These findings suggest that rapid proBDNF increases following SE are due in part to reduced cleavage, and that proBDNF may be part of the initial neurotrophin response driving intracellular signaling during the acute phase of epileptogenesis.


Subject(s)
Brain-Derived Neurotrophic Factor/metabolism , Hippocampus/metabolism , Plasminogen Activator Inhibitor 1/metabolism , Protein Precursors/metabolism , Protein Processing, Post-Translational , Status Epilepticus/metabolism , Animals , Astrocytes/metabolism , Male , Mice , Mice, Inbred C57BL , Neurons/metabolism , Pilocarpine , Status Epilepticus/chemically induced
15.
Article in English | MEDLINE | ID: mdl-26330656

ABSTRACT

Brain-derived neurotrophic factor (BDNF) is a member of a family of neurotrophins which include nerve growth factor, neurotrophin 3, and neurotrophin 4. Studies over the last three decades have identified mature BDNF as a key regulator of neuronal differentiation, structure, and function; actions mediated by the TrkB receptor. More recently identified isoforms which are translated from the bdnf gene, including the uncleaved precursor, pro-BDNF, and the cleaved prodomain, have been found to elicit opposing functions in neurons through the activation of distinct receptors. This work emphasizes the critical roles for all three isoforms of BDNF in modulating neuronal activity that impact complex human behaviors including memory, anxiety, depression, and hyperphagia.


Subject(s)
Brain-Derived Neurotrophic Factor/metabolism , Receptor, trkB/metabolism , Signal Transduction , Animals , Brain-Derived Neurotrophic Factor/chemistry , Brain-Derived Neurotrophic Factor/genetics , Gene Expression Regulation , Humans , Ligands , Polymorphism, Genetic , Protein Conformation , Protein Isoforms , Structure-Activity Relationship
16.
J Neurosci ; 35(34): 11911-20, 2015 Aug 26.
Article in English | MEDLINE | ID: mdl-26311773

ABSTRACT

The p75 neurotrophin receptor (p75(NTR)) is a multifunctional receptor that participates in many critical processes in the nervous system, ranging from apoptosis to synaptic plasticity and morphological events. It is a member of the tumor necrosis factor receptor (TNFR) superfamily, whose members undergo trimeric oligomerization. Interestingly, p75(NTR) interacts with dimeric ligands (i.e., proneurotrophins or mature neurotrophins), but several of the intracellular adaptors that mediate p75(NTR) signaling are trimeric (i.e., TNFR-associated factor 6 or TRAF6). Consequently, the active receptor signaling unit remains uncertain. To identify the functional receptor complex, we evaluated its oligomerization in vitro and in mice brain tissues using a combination of biochemical techniques. We found that the most abundant homotypic arrangement for p75(NTR) is a trimer and that monomers and trimers coexist at the cell surface. Interestingly, trimers are not required for ligand-independent or ligand-dependent p75(NTR) activation in a growth cone retraction functional assay. However, monomers are capable of inducing acute morphological effects in neurons. We propose that p75(NTR) activation is regulated by its oligomerization status and its levels of expression. These results indicate that the oligomeric state of p75(NTR) confers differential responses and offers an explanation for the diverse and contradictory actions of this receptor in the nervous system. SIGNIFICANCE STATEMENT: The p75 neurotrophin receptor (p75(NTR)) regulates a wide range of cellular functions, including apoptosis, neuronal processes remodeling, and synaptic plasticity. The goal of our work was to inquire whether oligomers of the receptor are required for function. Here we report that p75(NTR) predominantly assembles as a trimer, similar to other tumor necrosis factor receptors. Interestingly, monomers and trimers coexist at the cell surface, but trimers are not required for p75(NTR) activation in a functional assay. However, monomers are capable of inducing acute morphological effects in neurons. Identification of the oligomerization state of p75(NTR) begins to provide insights to the mechanisms of signal initiation of this noncatalytic receptor, as well as to develop therapeutic interventions to diminish its activity.


Subject(s)
Nerve Tissue Proteins/biosynthesis , Nerve Tissue Proteins/chemistry , Receptors, Nerve Growth Factor/biosynthesis , Receptors, Nerve Growth Factor/chemistry , Animals , Cells, Cultured , Cerebral Cortex/metabolism , Female , HEK293 Cells , Hippocampus/metabolism , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Nerve Tissue Proteins/genetics , PC12 Cells , Rats , Receptors, Nerve Growth Factor/genetics , Stereoisomerism
17.
Neurobiol Dis ; 82: 466-477, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26282324

ABSTRACT

Huntington's disease (HD) is a neurodegenerative disorder characterized by massive loss of medium spiny neurons in the striatum. However, the mechanisms by which mutant huntingtin leads to this selective neuronal death remain incompletely understood. Brain-derived neurotrophic factor (BDNF) has been shown to be neuroprotective on HD striatal neurons both in vitro and in vivo. ProBDNF, the precursor of mature BDNF (mBDNF), also can be secreted but promotes apoptosis of neurons expressing p75(NTR) and sortilin receptors. Although a reduction of total striatal BDNF protein has been reported in HD patients and mouse models, it remains unclear whether conversion of proBDNF to mBDNF is altered in HD, and whether the proBDNF receptors, p75(NTR) and sortilin are dysregulated, leading to impaired striatal neuron survival. To test these hypotheses, we generated bdnf-HA knock-in (KI) mice on the zQ175 HD background to accurately quantitate the levels of both proBDNF and mBDNF in the HD striatum. In aged zQ175 HD mice, we observed a significant loss of mBDNF and decreased TrkB activation, but no increase of proBDNF or p75(NTR) levels either in the sensorimotor cortex or the striatum. However, immunoreactivities of p75(NTR) and sortilin receptor are both increased in immature striatal oligodendrocytes, which associate with significant myelin defects in the HD striatum. Taken together, the present study indicates that diminished mature BDNF trophic signaling through the TrkB receptor, rather than an induction in proBDNF, is a main contributing factor to the vulnerability of striatal neurons in the zQ175 HD mouse model.


Subject(s)
Aging/metabolism , Brain-Derived Neurotrophic Factor/metabolism , Corpus Striatum/metabolism , Hippocampus/metabolism , Huntington Disease/metabolism , Sensorimotor Cortex/metabolism , Adaptor Proteins, Vesicular Transport/metabolism , Aging/pathology , Animals , Corpus Striatum/ultrastructure , Disease Models, Animal , Gene Knock-In Techniques , Huntington Disease/pathology , Male , Mice, Inbred C57BL , Mice, Transgenic , Myelin Sheath/metabolism , Myelin Sheath/ultrastructure , Oligodendroglia/metabolism , Oligodendroglia/ultrastructure , Protein Isoforms , Receptor, trkB/metabolism , Receptors, Nerve Growth Factor/metabolism
18.
Dev Cell ; 33(6): 690-702, 2015 Jun 22.
Article in English | MEDLINE | ID: mdl-26004511

ABSTRACT

Recent studies in humans and in genetic mouse models have identified Slit- and NTRK-like family (Slitrks) as candidate genes for neuropsychiatric disorders. All Slitrk isotypes are highly expressed in the CNS, where they mediate neurite outgrowth, synaptogenesis, and neuronal survival. However, the molecular mechanisms underlying these functions are not known. Here, we report that Slitrk5 modulates brain-derived neurotrophic factor (BDNF)-dependent biological responses through direct interaction with TrkB receptors. Under basal conditions, Slitrk5 interacts primarily with a transsynaptic binding partner, protein tyrosine phosphatase δ (PTPδ); however, upon BDNF stimulation, Slitrk5 shifts to cis-interactions with TrkB. In the absence of Slitrk5, TrkB has a reduced rate of ligand-dependent recycling and altered responsiveness to BDNF treatment. Structured illumination microscopy revealed that Slitrk5 mediates optimal targeting of TrkB receptors to Rab11-positive recycling endosomes through recruitment of a Rab11 effector protein, Rab11-FIP3. Thus, Slitrk5 acts as a TrkB co-receptor that mediates its BDNF-dependent trafficking and signaling.


Subject(s)
Brain-Derived Neurotrophic Factor/metabolism , Membrane Proteins/metabolism , Nerve Tissue Proteins/metabolism , Receptor, trkB/metabolism , Animals , Corpus Striatum/metabolism , Endosomes/metabolism , HEK293 Cells , Humans , Membrane Proteins/deficiency , Membrane Proteins/genetics , Mice , Mice, Knockout , Nerve Tissue Proteins/deficiency , Nerve Tissue Proteins/genetics , Neurons/metabolism , Protein Binding , Protein Transport , Receptor-Like Protein Tyrosine Phosphatases, Class 2/metabolism , Signal Transduction , rab GTP-Binding Proteins/metabolism
19.
Biomaterials ; 55: 110-8, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25934456

ABSTRACT

Oxygen status and tissue dimensionality are critical determinants of tumor angiogenesis, a hallmark of cancer and an enduring target for therapeutic intervention. However, it is unclear how these microenvironmental conditions interact to promote neovascularization, due in part to a lack of comprehensive, unbiased data sets describing tumor cell gene expression as a function of oxygen levels within three-dimensional (3D) culture. Here, we utilized alginate-based, oxygen-controlled 3D tumor models to study the interdependence of culture context and the hypoxia response. Microarray gene expression analysis of tumor cells cultured in 2D versus 3D under ambient or hypoxic conditions revealed striking interdependence between culture dimensionality and hypoxia response, which was mediated in part by pro-inflammatory signaling pathways. In particular, interleukin-8 (IL-8) emerged as a major player in the microenvironmental regulation of the hypoxia program. Notably, this interaction between dimensionality and oxygen status via IL-8 increased angiogenic sprouting in a 3D endothelial invasion assay. Taken together, our data suggest that pro-inflammatory pathways are critical regulators of tumor hypoxia response within 3D environments that ultimately impact tumor angiogenesis, potentially providing important therapeutic targets. Furthermore, these results highlight the importance of pathologically relevant tissue culture models to study the complex physical and chemical processes by which the cancer microenvironment mediates new vessel formation.


Subject(s)
Biocompatible Materials/chemistry , Cell Culture Techniques , Neoplasms/pathology , Neovascularization, Pathologic , Oxygen/chemistry , Alginates/chemistry , Cell Hypoxia , Endothelium, Vascular/pathology , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Glucuronic Acid/chemistry , Hexuronic Acids/chemistry , Human Umbilical Vein Endothelial Cells , Humans , Hydrogels/chemistry , Inflammation/pathology , Interleukin-8/metabolism , Neoplasm Invasiveness , Oligonucleotide Array Sequence Analysis , Signal Transduction , Tissue Engineering/methods , Tumor Cells, Cultured
20.
Nicotine Tob Res ; 17(12): 1428-35, 2015 Dec.
Article in English | MEDLINE | ID: mdl-25744957

ABSTRACT

INTRODUCTION: Nicotine withdrawal is characterized by both affective and cognitive symptoms. Identifying genetic polymorphisms that could affect the symptoms associated with nicotine withdrawal are important in predicting withdrawal sensitivity and identifying personalized cessation therapies. In the current study we used a mouse model of a non-synonymous single nucleotide polymorphism in the translated region of the brain-derived neurotrophic factor (BDNF) gene that substitutes a valine (Val) for a methionine (Met) amino acid (Val66Met) to examine the relationship between the Val66Met single nucleotide polymorphism and nicotine dependence. METHODS: This study measured proBDNF and the BDNF prodomain levels following nicotine and nicotine withdrawal and examined a mouse model of a common polymorphism in this protein (BDNF(Met/Met)) in three behavioral paradigms: novelty-induced hypophagia, marble burying, and the open-field test. RESULTS: Using the BDNF knock-in mouse containing the BDNF Val66Met polymorphism we found: (1) blunted anxiety-like behavior in BDNF(Met/Met) mice following withdrawal in three behavioral paradigms: novelty-induced hypophagia, marble burying, and the open-field test; (2) the anxiolytic effects of chronic nicotine are absent in BDNF(Met/Met) mice; and (3) an increase in BDNF prodomain in BDNF(Met/Met) mice following nicotine withdrawal. CONCLUSIONS: Our study is the first to examine the effect of the BDNF Val66Met polymorphism on the affective symptoms of withdrawal from nicotine in mice. In these mice, a single-nucleotide polymorphism in the translated region of the BDNF gene can result in a blunted withdrawal, as measured by decreased anxiety-like behavior. The significant increase in the BDNF prodomain in BDNF(Met/Met) mice following nicotine cessation suggests a possible role of this ligand in the circuitry remodeling after withdrawal.


Subject(s)
Anxiety/genetics , Brain-Derived Neurotrophic Factor/genetics , Methionine/genetics , Nicotine/administration & dosage , Substance Withdrawal Syndrome/genetics , Valine/genetics , Animals , Female , Gene Knock-In Techniques , Male , Mice , Mice, Transgenic , Polymorphism, Single Nucleotide/genetics , Substance Withdrawal Syndrome/psychology , Tobacco Use Disorder/genetics , Tobacco Use Disorder/psychology
SELECTION OF CITATIONS
SEARCH DETAIL
...