Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 457
Filter
1.
J Neurosci ; 41(18): 3948-3957, 2021 05 05.
Article in English | MEDLINE | ID: mdl-33789918

ABSTRACT

Drosophila odorant receptors (Ors) are ligand gated ion channels composed of a common receptor subunit Or co-receptor (ORCO) and one of 62 "tuning" receptor subunits that confer odorant specificity to olfactory neuron responses. Like other sensory systems studied to date, exposing Drosophila olfactory neurons to activating ligands results in reduced responses to subsequent exposures through a process called desensitization. We recently showed that phosphorylation of serine 289 on the common Or subunit ORCO is required for normal peak olfactory neuron responses. Dephosphorylation of this residue occurs on prolonged odorant exposure, and underlies the slow modulation of olfactory neuron responses we term "slow desensitization." Slow desensitization results in the reduction of peak olfactory neuron responses and flattening of dose-response curves, implicating changes in ORCOS289 phosphorylation state as an important modulator of olfactory neuron responses. Here, we report the identification of the primary kinase responsible for ORCOS289 phosphorylation, PKC98E. Antiserum localizes the kinase to the dendrites of the olfactory neurons. Deletion of the kinase from olfactory neurons in the naive state (the absence of prolonged odor exposure) reduces ORCOS289 phosphorylation and reduces peak odorant responses without altering receptor localization or expression levels. Genetic rescue with a PKC98E predicted to be constitutively active restores ORCO S289 phosphorylation and olfactory neuron sensitivity to the PKC98E mutants in the naive state. However, the dominant kinase is defective for slow desensitization. Together, these findings reveal that PKC98E is an important regulator of ORCO receptors and olfactory neuron function.SIGNIFICANCE STATEMENT We have identified PKC98E as the kinase responsible for phosphorylation of the odorant receptor co-receptor (ORCO) at S289 that is required for normal odorant response kinetics of olfactory neurons. This is a significant step toward revealing the enzymology underlying the regulation of odorant response regulation in insects.


Subject(s)
Drosophila Proteins/physiology , Drosophila melanogaster/physiology , Olfactory Receptor Neurons/physiology , Protein Kinase C/physiology , Animals , Dendrites/enzymology , Dose-Response Relationship, Drug , Drosophila Proteins/genetics , Electrophysiological Phenomena , Gene Deletion , Mutation/genetics , Odorants , Olfactory Receptor Neurons/enzymology , Phosphorylation , Protein Kinase C/genetics , RNA Interference , Receptors, Odorant/genetics , Receptors, Odorant/metabolism
2.
Int J Mol Sci ; 21(15)2020 Jul 28.
Article in English | MEDLINE | ID: mdl-32731326

ABSTRACT

Skin hyperpigmentation is generally characterized by increased synthesis and deposition of melanin in the skin. UP256, containing bakuchiol, is a well-known medication for acne vulgaris. Acne sometimes leaves dark spots on the skin, and we hypothesized that UP256 may be effective against hyperpigmentation-associated diseases. UP256 was treated for anti-melanogenesis and melanocyte dendrite formation in cultured normal human epidermal melanocytes as well as in the reconstituted skin and zebrafish models. Western blot analysis and glutathione S-transferase (GST)-pull down assays were used to evaluate the expression and interaction of enzymes related in melanin synthesis and transportation. The cellular tyrosinase activity and melanin content assay revealed that UP256 decreased melanin synthesis by regulating the expression of proteins related on melanogenesis including tyrosinase, TRP-1 and -2, and SOX9. UP256 also decreased dendrite formation in melanocytes via regulating the Rac/Cdc42/α-PAK signaling proteins, without cytotoxic effects. UP256 also inhibited ciliogenesis-dependent melanogenesis in normal human epidermal melanocytes. Furthermore, UP256 suppressed melanin contents in the zebrafish and the 3D human skin tissue model. All things taken together, UP256 inhibits melanin synthesis, dendrite formation, and primary cilium formation leading to the inhibition of melanogenesis.


Subject(s)
Cilia/enzymology , Gene Expression Regulation, Enzymologic , Hyperpigmentation/enzymology , Melanocytes/enzymology , Monophenol Monooxygenase/biosynthesis , Signal Transduction , Up-Regulation , Zebrafish Proteins/biosynthesis , Zebrafish/metabolism , Animals , Cell Line , Cilia/pathology , Dendrites/enzymology , Dendrites/pathology , Humans , Hyperpigmentation/drug therapy , Hyperpigmentation/pathology , SOX9 Transcription Factor/metabolism , Trypsin/metabolism
3.
Mol Psychiatry ; 25(5): 1022-1034, 2020 05.
Article in English | MEDLINE | ID: mdl-30120419

ABSTRACT

Depression alters the structure and function of brain reward circuitry. Preclinical evidence suggests that medium spiny neurons (MSNs) in the nucleus accumbens (NAc) undergo structural plasticity; however, the molecular mechanism and behavioral significance is poorly understood. Here we report that atrophy of D1, but not D2 receptor containing MSNs is strongly associated with social avoidance in mice subject to social defeat stress. D1-MSN atrophy is caused by cell-type specific upregulation of the GTPase RhoA and its effector Rho-kinase. Pharmacologic and genetic reduction of activated RhoA prevents depressive outcomes to stress by preventing loss of D1-MSN dendritic arbor. Pharmacologic and genetic promotion of activated RhoA enhances depressive outcomes by reducing D1-MSN dendritic arbor and is sufficient to promote depressive-like behaviors in the absence of stress. Chronic treatment with Rho-kinase inhibitor Y-27632 after chronic social defeat stress reverses depression-like behaviors by restoring D1-MSN dendritic complexity. Taken together, our data indicate functional roles for RhoA and Rho-kinase in mediating depression-like behaviors via dendritic remodeling of NAc D1-MSNs and may prove a useful target for new depression therapeutics.


Subject(s)
Dendrites/enzymology , Dendrites/pathology , Depression/pathology , Depression/psychology , Neuronal Plasticity , Receptors, Dopamine D1/metabolism , rho-Associated Kinases/metabolism , rhoA GTP-Binding Protein/metabolism , Animals , Depression/enzymology , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Nucleus Accumbens/metabolism , Nucleus Accumbens/pathology , Receptors, Dopamine D2/metabolism
4.
Elife ; 82019 10 28.
Article in English | MEDLINE | ID: mdl-31657720

ABSTRACT

Parvalbumin-expressing (PV+) GABAergic interneurons mediate feedforward and feedback inhibition and have a key role in gamma oscillations and information processing. The importance of fast synaptic recruitment and action potential initiation and repolarization, and rapid synchronous GABA release by PV+ cells, is well established. In contrast, the functional significance of PV+ cell NMDA receptors (NMDARs), which generate relatively slow postsynaptic currents, is unclear. Underlining their potential importance, several studies implicate PV+ cell NMDAR disruption in impaired network function and circuit pathologies. Here, we show that dendritic NMDARs underlie supralinear integration of feedback excitation from local pyramidal neurons onto mouse CA1 PV+ cells. Furthermore, by incorporating NMDARs at feedback connections onto PV+ cells in spiking networks, we show that these receptors enable cooperative recruitment of PV+ interneurons, strengthening and stabilising principal cell assemblies. Failure of this phenomenon provides a parsimonious explanation for cognitive and sensory gating deficits in pathologies with impaired PV+ NMDAR signalling.


Subject(s)
Dendrites/physiology , GABAergic Neurons/physiology , Interneurons/physiology , Nerve Net/physiology , Pyramidal Cells/physiology , Receptors, N-Methyl-D-Aspartate/metabolism , Action Potentials , Animals , CA1 Region, Hippocampal/cytology , CA1 Region, Hippocampal/physiology , Dendrites/enzymology , GABAergic Neurons/enzymology , Interneurons/enzymology , Mice
5.
J Tissue Eng Regen Med ; 13(1): 110-115, 2019 01.
Article in English | MEDLINE | ID: mdl-30479062

ABSTRACT

Amyotrophic lateral sclerosis (ALS) is a fatal neurodegenerative disease affecting motor neuron system. Our previous study has shown that bone marrow-mesenchymal stem cells (BM-MSCs) from ALS patients have functional limitations in releasing neurotrophic factors and exhibit the senescence phenotype. In this study, we examined sirtuin 1/adenosine monophosphate-activated protein kinase (SIRT1/AMPK) activities and identified significant decreases in the ALS-MSCs compared with normal healthy control originated BM-MSCs. This decline was restored by pretreatment with resveratrol (RSV), measured using quantitative polymerase chain reaction, NAD/NADH assay, and immunoblot analysis. Neuroprogenitor markers were increased in RSV-treated ALS-MSCs (RSV/ALS-MSCs). The differentiated ALS-MSCs (ALS-dMSCs) exhibited a cell body and dendritic shape similar to neurons. RSV/ALS-MSCs showed significantly increased differentiation rate as compared with the untreated ALS-dMSCs. The neurite numbers and lengths were also significantly increased. This was confirmed with immunoblot analysis using neuron specific markers such as nestin, NF-M, Tuj-1, and Map-2 in RSV/ALS-dMSCs. Thus, this study shows that ALS-MSCs showed down-regulation of AMPK/SIRT1 signalling, which was recovered by treatment with RSV. This data suggest that RSV can be one of the candidate agents for improving therapeutic efficacy of ALS patients' originated MSCs.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Amyotrophic Lateral Sclerosis/enzymology , Cell Differentiation/drug effects , Mesenchymal Stem Cells/enzymology , Resveratrol/pharmacology , Sirtuin 1/biosynthesis , Amyotrophic Lateral Sclerosis/pathology , Dendrites/enzymology , Female , Humans , Male , Mesenchymal Stem Cells/pathology
6.
J Cell Biol ; 217(10): 3577-3592, 2018 10 01.
Article in English | MEDLINE | ID: mdl-30087126

ABSTRACT

INPP5K (SKIP) is an inositol 5-phosphatase that localizes in part to the endoplasmic reticulum (ER). We show that recruitment of INPP5K to the ER is mediated by ARL6IP1, which shares features of ER-shaping proteins. Like ARL6IP1, INPP5K is preferentially localized in ER tubules and enriched, relative to other ER resident proteins (Sec61ß, VAPB, and Sac1), in newly formed tubules that grow along microtubule tracks. Depletion of either INPP5K or ARL6IP1 results in the increase of ER sheets. In a convergent but independent study, a screen for mutations affecting the distribution of the ER network in dendrites of the PVD neurons of Caenorhabditis elegans led to the isolation of mutants in CIL-1, which encodes the INPP5K worm orthologue. The mutant phenotype was rescued by expression of wild type, but not of catalytically inactive CIL-1. Our results reveal an unexpected role of an ER localized polyphosphoinositide phosphatase in the fine control of ER network organization.


Subject(s)
Caenorhabditis elegans Proteins/metabolism , Caenorhabditis elegans/enzymology , Dendrites/enzymology , Endoplasmic Reticulum/enzymology , Phosphatidylinositol-3,4,5-Trisphosphate 5-Phosphatases/metabolism , Animals , COS Cells , Caenorhabditis elegans/genetics , Caenorhabditis elegans Proteins/genetics , Chlorocebus aethiops , Dendrites/genetics , Endoplasmic Reticulum/genetics , Gene Deletion , HeLa Cells , Humans , Mice , Phosphatidylinositol-3,4,5-Trisphosphate 5-Phosphatases/genetics
7.
Methods Mol Biol ; 1821: 297-317, 2018.
Article in English | MEDLINE | ID: mdl-30062421

ABSTRACT

Most neurons elaborate a characteristic dendritic arbor which is physiologically important for receiving and processing of synaptic inputs. Pathologically, disturbances in the regulation of dendritic tree complexity are often associated with mental retardation and other neurological deficits. Rho GTPases are major players in the regulation of dendritic tree complexity. They are involved in many signal transduction cascades, activated at the neuronal plasma membrane, and relayed to intracellular proteins that directly rearrange the cytoskeleton. The use of siRNA technology combined with morphometric and imaging techniques allows the roles of individual Rho GTPases, such as Rac1, in dendritic branching to be examined. In this chapter we describe the establishment, transfection, and processing of a primary hippocampal cell culture. Methods to assess the complexity of dendritic arbors like the Sholl analysis, and techniques to investigate Rac1 activity in hippocampal cells, and specifically in neuronal dendrites, such as fluorescence resonance energy transfer (FRET) imaging are presented.


Subject(s)
Dendrites/enzymology , Gene Silencing , Hippocampus/enzymology , Nerve Net/enzymology , RNA, Small Interfering , Transfection/methods , rac1 GTP-Binding Protein , Animals , Hippocampus/cytology , Nerve Net/cytology , RNA, Small Interfering/biosynthesis , RNA, Small Interfering/genetics , Rats , Rats, Wistar , rac1 GTP-Binding Protein/antagonists & inhibitors , rac1 GTP-Binding Protein/biosynthesis , rac1 GTP-Binding Protein/genetics
8.
J Neurosci ; 38(35): 7701-7712, 2018 08 29.
Article in English | MEDLINE | ID: mdl-30030398

ABSTRACT

In addition to mechanisms promoting protein-synthesis-dependent long-term memory (PSD-LTM), the process appears to also be specifically constrained. We present evidence that the highly conserved receptor tyrosine kinase dAlk is a novel PSD-LTM attenuator in Drosophila Reduction of dAlk levels in adult α/ß mushroom body (MB) neurons during conditioning elevates LTM, whereas its overexpression impairs it. Unlike other memory suppressor proteins and miRNAs, dAlk within the MBs constrains PSD-LTM specifically but constrains learning outside the MBs as previously shown. Dendritic dAlk levels rise rapidly in MB neurons upon conditioning, a process apparently controlled by the 3'UTR of its mRNA, and interruption of the 3'UTR leads to enhanced LTM. Because its activating ligand Jeb is dispensable for LTM attenuation, we propose that postconditioning elevation of dAlk within α/ß dendrites results in its autoactivation and constrains formation of the energy costly PSD-LTM, acting as a novel memory filter.SIGNIFICANCE STATEMENT In addition to the widely studied molecular mechanisms promoting protein-synthesis-dependent long-term memory (PSD-LTM), recent discoveries indicate that the process is also specifically constrained. We describe a role in PSD-LTM constraint for the first receptor tyrosine kinase (RTK) involved in olfactory memory in Drosophila Unlike other memory suppressor proteins and miRNAs, dAlk limits specifically PSD-LTM formation as it does not affect 3 h, or anesthesia-resistant memory. Significantly, we show conditioning-dependent dAlk elevation within the mushroom body dendrites and propose that its local abundance may activate its kinase activity, to mediate imposition of PSD-LTM constraints through yet unknown mechanisms.


Subject(s)
Anaplastic Lymphoma Kinase/physiology , Avoidance Learning/physiology , Drosophila Proteins/physiology , Drosophila/physiology , Memory, Long-Term/physiology , Nerve Tissue Proteins/physiology , 3' Untranslated Regions , Anaplastic Lymphoma Kinase/antagonists & inhibitors , Anaplastic Lymphoma Kinase/genetics , Animals , Dendrites/enzymology , Dendrites/physiology , Drosophila/enzymology , Drosophila/genetics , Drosophila/growth & development , Drosophila Proteins/antagonists & inhibitors , Drosophila Proteins/genetics , Enzyme Induction , Larva , Memory Consolidation , Mushroom Bodies/physiology , Nerve Tissue Proteins/biosynthesis , Neurons/physiology , Odorants , Pyrimidines/pharmacology , RNA Interference
9.
J Cell Sci ; 131(13)2018 07 09.
Article in English | MEDLINE | ID: mdl-29898920

ABSTRACT

The formation of complex dendritic arbors is crucial for the assembly of functional networks as abnormal dendrite formation underlies several neurodevelopmental and psychiatric disorders. Many extracellular factors have been postulated as regulators of dendritic growth. Wnt proteins play a critical role in neuronal development and circuit formation. We previously demonstrated that Wnt7b acts through the scaffold protein dishevelled 1 (Dvl1) to modulate dendrite arborisation by activating a non-canonical Wnt signalling pathway. Here, we identify the seven-transmembrane frizzled-7 (Fz7, also known as FZD7) as the receptor for Wnt7b-mediated dendrite growth and complexity. Importantly, Fz7 is developmentally regulated in the intact hippocampus, and is localised along neurites and at dendritic growth cones, suggesting a role in dendrite formation and maturation. Fz7 loss-of-function studies demonstrated that Wnt7b requires Fz7 to promote dendritic arborisation. Moreover, in vivo Fz7 loss of function results in dendritic defects in the intact mouse hippocampus. Furthermore, our findings reveal that Wnt7b and Fz7 induce the phosphorylation of Ca2+/calmodulin-dependent protein kinase II (CaMKII) and JNK proteins, which are required for dendritic development. Here, we demonstrate that Wnt7b-Fz7 signals through two non-canonical Wnt pathways to modulate dendritic growth and complexity.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Dendrites/metabolism , Hippocampus/growth & development , MAP Kinase Kinase 4/metabolism , Proto-Oncogene Proteins/metabolism , Receptors, G-Protein-Coupled/metabolism , Wnt Proteins/metabolism , Animals , Calcium-Calmodulin-Dependent Protein Kinase Type 2/genetics , Dendrites/enzymology , Dendrites/genetics , Dishevelled Proteins/genetics , Dishevelled Proteins/metabolism , Frizzled Receptors , Hippocampus/metabolism , MAP Kinase Kinase 4/genetics , Mice , Mice, Inbred C57BL , Neurites/metabolism , Protein Binding , Proto-Oncogene Proteins/genetics , Rats , Rats, Wistar , Receptors, G-Protein-Coupled/genetics , Wnt Proteins/genetics , Wnt Signaling Pathway
10.
Genes Dev ; 32(5-6): 402-414, 2018 03 01.
Article in English | MEDLINE | ID: mdl-29563183

ABSTRACT

Dendrites possess distinct structural and functional properties that enable neurons to receive information from the environment as well as other neurons. Despite their key role in neuronal function, current understanding of the ability of neurons to regenerate dendrites is lacking. This study characterizes the structural and functional capacity for dendrite regeneration in vivo in adult animals and examines the effect of neuronal maturation on dendrite regeneration. We focused on the class IV dendritic arborization (c4da) neuron of the Drosophila sensory system, which has a dendritic arbor that undergoes dramatic remodeling during the first 3 d of adult life and then maintains a relatively stable morphology thereafter. Using a laser severing paradigm, we monitored regeneration after acute and spatially restricted injury. We found that the capacity for regeneration was present in adult neurons but diminished as the animal aged. Regenerated dendrites recovered receptive function. Furthermore, we found that the regenerated dendrites show preferential alignment with the extracellular matrix (ECM). Finally, inhibition of ECM degradation by inhibition of matrix metalloproteinase 2 (Mmp2) to preserve the extracellular environment characteristics of young adults led to increased dendrite regeneration. These results demonstrate that dendrites retain regenerative potential throughout adulthood and that regenerative capacity decreases with aging.


Subject(s)
Dendrites/physiology , Drosophila/physiology , Matrix Metalloproteinase 2/metabolism , Regeneration , Sensory Receptor Cells/physiology , Aging/physiology , Animals , Dendrites/enzymology , Drosophila/cytology , Drosophila/enzymology , Drosophila Proteins/metabolism , Epidermis/enzymology , Extracellular Matrix/physiology , Gene Expression Regulation, Developmental , Integrins/genetics , Integrins/metabolism , Sensory Receptor Cells/enzymology
11.
J Comp Neurol ; 526(4): 742-766, 2018 03 01.
Article in English | MEDLINE | ID: mdl-29218725

ABSTRACT

We report the retinal expression pattern of Ret, a receptor tyrosine kinase for the glial derived neurotrophic factor (GDNF) family ligands (GFLs), during development and in the adult mouse. Ret is initially expressed in retinal ganglion cells (RGCs), followed by horizontal cells (HCs) and amacrine cells (ACs), beginning with the early stages of postmitotic development. Ret expression persists in all three classes of neurons in the adult. Using RNA sequencing, immunostaining and random sparse recombination, we show that Ret is expressed in at least three distinct types of ACs, and ten types of RGCs. Using intersectional genetics, we describe the dendritic arbor morphologies of RGC types expressing Ret in combination with each of the three members of the POU4f/Brn3 family of transcription factors. Ret expression overlaps with Brn3a in 4 RGC types, with Brn3b in 5 RGC types, and with Brn3c in one RGC type, respectively. Ret+ RGCs project to the lateral geniculate nucleus (LGN), pretectal area (PTA) and superior colliculus (SC), and avoid the suprachiasmatic nucleus and accessory optic system. Brn3a+ Ret+ and Brn3c+ Ret+ RGCs project preferentially to contralateral retinorecipient areas, while Brn3b+ Ret+ RGCs shows minor ipsilateral projections to the olivary pretectal nucleus and the LGN. Our findings establish intersectional genetic approaches for the anatomic and developmental characterization of individual Ret+ RGC types. In addition, they provide necessary information for addressing the potential interplay between GDNF neurotrophic signaling and transcriptional regulation in RGC type specification.


Subject(s)
Amacrine Cells/enzymology , Proto-Oncogene Proteins c-ret/metabolism , Retinal Ganglion Cells/enzymology , Retinal Horizontal Cells/enzymology , Amacrine Cells/cytology , Animals , Dendrites/enzymology , Gene Expression Profiling , Gene Expression Regulation, Developmental , Immunohistochemistry , Mice, Transgenic , Retinal Ganglion Cells/cytology , Retinal Horizontal Cells/cytology , Transcription Factor Brn-3A/metabolism , Visual Pathways/cytology , Visual Pathways/enzymology , Visual Pathways/growth & development
12.
Mol Cell Neurosci ; 85: 226-234, 2017 12.
Article in English | MEDLINE | ID: mdl-29107098

ABSTRACT

Stressor exposure induces neuronal remodeling in specific brain regions. Given the persistence of stress-related illnesses, key next steps in determining the contributions of neural structure to mental health are to identify cell types that fail to recover from stressor exposure and to identify "trigger points" and molecular underpinnings of stress-related neural degeneration. We evaluated dendrite arbor structure on hippocampal CA1 pyramidal neurons before, during, and following prolonged exposure to one key mediator of the stress response - corticosterone (cortisol in humans). Basal dendrite arbors progressively simplified during a 3-week exposure period, and failed to recover when corticosterone was withdrawn. Corticosterone exposure decreased levels of the dendrite stabilization factor Abl2/Arg nonreceptor tyrosine kinase and phosphorylation of its substrates p190RhoGAP and cortactin within 11days, suggesting that disruption of Arg-mediated signaling may trigger dendrite arbor atrophy and, potentially, behavioral abnormalities resulting from corticosterone exposure. To test this, we administered the novel, bioactive Arg kinase activator, 5-(1,3-diaryl-1H-pyrazol-4-yl)hydantoin, 5-[3-(4-fluorophenyl)-1-phenyl-1H-pyrazol-4-yl]-2,4-imidazolidinedione (DPH), in conjunction with corticosterone. We found that repeated treatment corrected CA1 arbor structure, otherwise simplified by corticosterone. DPH also corrected corticosterone-induced errors in a hippocampal-dependent reversal learning task and anhedonic-like behavior. Thus, pharmacological compounds that target cytoskeletal regulators, rather than classical neurotransmitter systems, may interfere with stress-associated cognitive decline and mental health concerns.


Subject(s)
Corticosterone/toxicity , Enzyme Activation/physiology , Protein-Tyrosine Kinases/metabolism , Pyramidal Cells/drug effects , Stress, Psychological/metabolism , Adrenal Cortex Hormones/toxicity , Animals , CA1 Region, Hippocampal/drug effects , CA1 Region, Hippocampal/enzymology , Dendrites/drug effects , Dendrites/enzymology , Dendrites/pathology , Mice , Mice, Inbred C57BL , Pyramidal Cells/enzymology , Stress, Psychological/pathology
13.
Xi Bao Yu Fen Zi Mian Yi Xue Za Zhi ; 33(9): 1177-1181, 2017 Sep.
Article in Chinese | MEDLINE | ID: mdl-29089072

ABSTRACT

Objective To explore the changes of cytochrome oxidase (COX) activity in the pre-Botzinger complex (pre-BotC) of the brainstem. Methods The double labeling of COX histochemistry and pre-BotC marker neurokinin-1 receptor (NK1R) nanogold-silver immunohistochemical staining was conducted to determine COX activity in the pre-BotC, especially within different subcellular structures of this nucleus. COX activity was semi-quantitatively analyzed. Results Under the light microscope, NK1R-immunoreactive (NK1R-ir) product was mainly distributed along the neuronal membrane, clearly outlining pre-BotC neurons. COX histochemical staining in brown was extensively expressed in the somata and processes of NK1R-ir neurons. Under the electron microscope, NK1R-ir gold particles were mainly distributed along the inner surface of the membrane of the somata and dendrites. The cytoplasm was also found labeled with NK1R-ir gold particles. The mitochondrial shape and distribution were different in different subcellular structures (somata, axon terminals, dendrites) of the pre-BotC. They were usually round or oval in the somata and axon terminals, whereas in the dendrites, slender elongated mitochondria were the most common. Tubular and vesicular cristae were more commonly visualized in the somata, but lamellar-oriented cristae were frequently encountered in the dendrites and axon terminals. The mitochondria appeared clustered together in the axon terminals, but in scattered distribution and close to the membrane in the dendrites except at synapses, where they were densely distributed and enlarged locally close to the postsynaptic membrane. The close link of the mitochondria with synapses indicated functional requirement that postsynaptic signal neurotransmission needs a large amount of ATP consumption. COX active product was expressed in the mitochondrial cristae, where different densities of the cristae represented different level of COX activity. The higher level of COX activity was evident in the axon terminals and dendrites than that in the somata, being significantly different. Conclusion Subcellular different regions in the pre-BotC function differently and need different energy metabolisms, thereby axon terminals and dendrites require higher COX activity than somata. In particular at synapses, mitochondria are densely localized with high COX activity. The present study provides a new approach by combination of COX histochemistry with immuno-electron microscopic techniques to detect regional COX activity in different subcellular structures of neurons.


Subject(s)
Brain Stem/enzymology , Electron Transport Complex IV/metabolism , Animals , Brain Stem/ultrastructure , Dendrites/enzymology , Immunohistochemistry , Microscopy, Immunoelectron , Presynaptic Terminals/enzymology , Rats , Rats, Sprague-Dawley , Receptors, Neurokinin-1/analysis
14.
Elife ; 62017 10 23.
Article in English | MEDLINE | ID: mdl-29058675

ABSTRACT

Small conductance calcium-activated potassium channels (SK channels) are present in spines and can be activated by backpropagating action potentials (APs). This suggests they may play a critical role in spike-timing dependent synaptic plasticity (STDP). Consistent with this idea, EPSPs in both cortical and hippocampal pyramidal neurons were suppressed by preceding APs in an SK-dependent manner. In cortical pyramidal neurons EPSP suppression by preceding APs depended on their precise timing as well as the distance of activated synapses from the soma, was dendritic in origin, and involved SK-dependent suppression of NMDA receptor activation. As a result SK channel activation by backpropagating APs gated STDP induction during low-frequency AP-EPSP pairing, with both LTP and LTD absent under control conditions but present after SK channel block. These findings indicate that activation of SK channels in spines by backpropagating APs plays a key role in regulating both EPSP amplitude and STDP induction.


Subject(s)
Action Potentials , Dendrites/enzymology , Dendrites/physiology , Excitatory Postsynaptic Potentials , Neuronal Plasticity , Small-Conductance Calcium-Activated Potassium Channels/metabolism , Spine/cytology , Animals , Female , Male , Rats, Wistar
15.
Genes Dev ; 31(10): 1054-1065, 2017 05 15.
Article in English | MEDLINE | ID: mdl-28637694

ABSTRACT

Proper function of the neural network results from the precise connections between axons and dendrites of presynaptic and postsynaptic neurons, respectively. In the Drosophila olfactory system, the dendrites of projection neurons (PNs) stereotypically target one of ∼50 glomeruli in the antennal lobe (AL), the primary olfactory center in the brain, and form synapses with the axons of olfactory receptor neurons (ORNs). Here, we show that Eph and Ephrin, the well-known axon guidance molecules, instruct the dendrodendritic segregation during the discrete olfactory map formation. The Eph receptor tyrosine kinase is highly expressed and localized in the glomeruli related to reproductive behavior in the developing AL. In one of the pheromone-sensing glomeruli (DA1), the Eph cell-autonomously regulates its dendrites to reside in a single glomerulus by interacting with Ephrins expressed in adjacent PN dendrites. Our data demonstrate that the trans interaction between dendritic Eph and Ephrin is essential for the PN dendritic boundary formation in the DA1 olfactory circuit, potentially enabling strict segregation of odor detection between pheromones and the other odors.


Subject(s)
Drosophila melanogaster/physiology , Receptor, EphA1/metabolism , Animals , Dendrites/enzymology , Dendrites/physiology , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Drosophila melanogaster/embryology , Drosophila melanogaster/enzymology , Drosophila melanogaster/genetics , Gene Expression Profiling , Gene Expression Regulation, Developmental , Gene Knockdown Techniques , Membrane Proteins/genetics , Membrane Proteins/metabolism , Olfactory Receptor Neurons/cytology , Olfactory Receptor Neurons/enzymology , Olfactory Receptor Neurons/physiology , RNA Interference , Receptor, EphA1/genetics
16.
Dev Neurobiol ; 77(10): 1175-1187, 2017 10.
Article in English | MEDLINE | ID: mdl-28589675

ABSTRACT

Cyclin-dependent kinase 5 (Cdk5) is recognized as a unique member among other Cdks due to its versatile roles in many biochemical processes in the nervous system. The proper development of neuronal dendrites is required for the formation of complex neural networks providing the physiological basis of various neuronal functions. We previously reported that sparse dendrites were observed on cultured Cdk5-null Purkinje cells and Purkinje cells in Wnt1cre -mediated Cdk5 conditional knockout (KO) mice. In the present study, we generated L7cre -mediated p35; p39 double KO (L7cre -p35f/f ; p39-/- ) mice whose Cdk5 activity was eliminated specifically in Purkinje cells of the developing cerebellum. Consequently, these mice exhibited defective Purkinje cell migration, motor coordination deficiency and a Purkinje dendritic abnormality similar to what we have observed before, suggesting that dendritic growth of Purkinje cells was cell-autonomous in vivo. We found that mixed and overlay cultures of WT cerebellar cells rescued the dendritic deficits in Cdk5-null Purkinje cells, however, indicating that Purkinje cell dendritic development was also supported by non-cell-autonomous factors. We then again rescued these abnormalities in vitro by applying exogenous brain-derived neurotrophic factor (BDNF). Based on the results from culture experiments, we attempted to rescue the developmental defects of Purkinje cells in L7cre -p35f/f ; p39-/- mice by using a TrkB agonist. We observed partial rescue of morphological defects of dendritic structures of Purkinje cells. These results suggest that Cdk5 activity is required for Purkinje cell dendritic growth in cell-autonomous and non-cell-autonomous manners. © 2017 Wiley Periodicals, Inc. Develop Neurobiol 77: 1175-1187, 2017.


Subject(s)
Cyclin-Dependent Kinase 5/metabolism , Dendrites/enzymology , Neuronal Outgrowth/physiology , Purkinje Cells/enzymology , Animals , Brain-Derived Neurotrophic Factor/metabolism , Calbindins/metabolism , Cells, Cultured , Cerebellum/drug effects , Cerebellum/enzymology , Cerebellum/growth & development , Cerebellum/pathology , Dendrites/drug effects , Dendrites/pathology , Forelimb/drug effects , Forelimb/physiopathology , Immunohistochemistry , Male , Mice, Inbred C57BL , Mice, Knockout , Motor Activity/drug effects , Motor Activity/physiology , Muscle Strength/drug effects , Muscle Strength/physiology , Neuronal Outgrowth/drug effects , Purkinje Cells/drug effects , Purkinje Cells/pathology , Receptor, trkB/agonists , Receptor, trkB/metabolism , Rotarod Performance Test
17.
J Biol Chem ; 292(15): 6402-6413, 2017 04 14.
Article in English | MEDLINE | ID: mdl-28264928

ABSTRACT

An improved understanding of the molecular mechanisms in synapse formation provides insight into both learning and memory and the etiology of neurodegenerative disorders. Coactivator-associated arginine methyltransferase 1 (CARM1) is a protein methyltransferase that negatively regulates synaptic gene expression and inhibits neuronal differentiation. Despite its regulatory function in neurons, little is known about the CARM1 cellular location and its role in dendritic maturation and synapse formation. Here, we examined the effects of CARM1 inhibition on dendritic spine and synapse morphology in the rat hippocampus. CARM1 was localized in hippocampal post-synapses, with immunocytochemistry and electron microscopy revealing co-localization of CARM1 with post-synaptic density (PSD)-95 protein, a post-synaptic marker. Specific siRNA-mediated suppression of CARM1 expression resulted in precocious dendritic maturation, with increased spine width and density at sites along dendrites and induction of mushroom-type spines. These changes were accompanied by a striking increase in the cluster size and number of key synaptic proteins, including N-methyl-d-aspartate receptor subunit 2B (NR2B) and PSD-95. Similarly, pharmacological inhibition of CARM1 activity with the CARM1-specific inhibitor AMI-1 significantly increased spine width and mushroom-type spines and also increased the cluster size and number of NR2B and cluster size of PSD-95. These results suggest that CARM1 is a post-synaptic protein that plays roles in dendritic maturation and synaptic formation and that spatiotemporal regulation of CARM1 activity modulates neuronal connectivity and improves synaptic dysfunction.


Subject(s)
Dendrites/enzymology , Hippocampus/enzymology , Post-Synaptic Density/enzymology , Protein-Arginine N-Methyltransferases/metabolism , Animals , Cells, Cultured , Disks Large Homolog 4 Protein , Hippocampus/cytology , Intracellular Signaling Peptides and Proteins/metabolism , Membrane Proteins/metabolism , Rats , Receptors, N-Methyl-D-Aspartate/metabolism , Spinal Cord/cytology , Spinal Cord/enzymology
18.
J Cell Physiol ; 232(12): 3677-3692, 2017 Dec.
Article in English | MEDLINE | ID: mdl-28160495

ABSTRACT

Dendrite arbor growth, or dendritogenesis, is choreographed by a diverse set of cues, including the NMDA receptor (NMDAR) subunits NR2A and NR2B. While NR1NR2B receptors are predominantly expressed in immature neurons and promote plasticity, NR1NR2A receptors are mainly expressed in mature neurons and induce circuit stability. How the different subunits regulate these processes is unclear, but this is likely related to the presence of their distinct C-terminal sequences that couple different signaling proteins. Calcium-calmodulin-dependent protein kinase II (CaMKII) is an interesting candidate as this protein can be activated by calcium influx through NMDARs. CaMKII triggers a series of biochemical signaling cascades, involving the phosphorylation of diverse targets. Among them, the activation of cAMP response element-binding protein (CREB-P) pathway triggers a plasticity-specific transcriptional program through unknown epigenetic mechanisms. Here, we found that dendritogenesis in hippocampal neurons is impaired by several well-characterized constructs (i.e., NR2B-RS/QD) and peptides (i.e., tatCN21) that specifically interfere with the recruitment and interaction of CaMKII with the NR2B C-terminal domain. Interestingly, we found that transduction of NR2AΔIN, a mutant NR2A construct with increased interaction to CaMKII, reactivates dendritogenesis in mature hippocampal neurons in vitro and in vivo. To gain insights into the signaling and epigenetic mechanisms underlying NMDAR-mediated dendritogenesis, we used immunofluorescence staining to detect CREB-P and acetylated lysine 27 of histone H3 (H3K27ac), an activation-associated histone tail mark. In contrast to control mature neurons, our data shows that activation of the NMDAR/CaMKII/ERK-P/CREB-P signaling axis in neurons expressing NR2AΔIN is not correlated with increased nuclear H3K27ac levels.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Cyclic AMP Response Element-Binding Protein/metabolism , Dendrites/enzymology , Hippocampus/enzymology , Histones/metabolism , Neurogenesis , Neuronal Plasticity , Receptors, N-Methyl-D-Aspartate/metabolism , Acetylation , Animals , Calcium-Calmodulin-Dependent Protein Kinase Type 2/antagonists & inhibitors , Calcium-Calmodulin-Dependent Protein Kinase Type 2/genetics , Cells, Cultured , Dendrites/drug effects , Gestational Age , Hippocampus/drug effects , Hippocampus/embryology , Mutation , Neurogenesis/drug effects , Neuronal Plasticity/drug effects , Peptides/pharmacology , Phenotype , Protein Binding , Protein Interaction Domains and Motifs , Protein Kinase Inhibitors/pharmacology , Protein Processing, Post-Translational , RNA Interference , Rats, Sprague-Dawley , Receptors, N-Methyl-D-Aspartate/genetics , Signal Transduction , Transfection
19.
Acta Neuropathol Commun ; 4: 29, 2016 Mar 31.
Article in English | MEDLINE | ID: mdl-27036709

ABSTRACT

The transmembrane protein assembly γ-secretase is a key protease in regulated intramembrane processing (RIP) of around 100 type-1 transmembrane proteins. Importantly, it has a pathological role in Alzheimer disease (AD) as it generates the neurotoxic amyloid ß-peptide from the amyloid precursor protein (APP). Studies on γ-secretase location are therefore crucial both from a biological and a therapeutic perspective. Despite several years of efforts in many laboratories, it is not clear where in the neuron γ-secretase exerts it's activities. Technical challenges include the fact that the active enzyme contains four protein components and that most subcellular compartments cannot be spatially resolved by traditional light microscopy. Here, we have used a powerful combination of the two nanoscopy techniques STORM and STED microscopy to visualize the location of γ-secretase in neurons using an active-site specific probe, with a focus on the synapse. We show that γ-secretase is present in both the pre-and postsynaptic compartments. We further show that the enzyme is enriched very close to the synaptic cleft in the postsynaptic membrane, as well as to NMDA receptors, demonstrating that γ-secretase is present in the postsynaptic plasma membrane. Importantly, the expression of γ-secretase increased in the pre- and postsynaptic compartments with the size of the synapse, suggesting a correlation between γ-secretase activity and synapse maturation. Thus, our data shows the synaptic location with high precision in three dimensions and settles the long-lasting debate on the synaptic location of γ-secretase.


Subject(s)
Amyloid Precursor Protein Secretases/metabolism , Axons/enzymology , Dendrites/enzymology , Neurons/cytology , Synapses/metabolism , Animals , Cells, Cultured , Disks Large Homolog 4 Protein , Embryo, Mammalian , Guanylate Kinases/metabolism , Hippocampus/cytology , Image Processing, Computer-Assisted , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Microscopy, Confocal , Microtubule-Associated Proteins/metabolism , Neuroimaging , Receptors, N-Methyl-D-Aspartate/metabolism , Synaptophysin/metabolism , tau Proteins/metabolism
20.
Brain Res ; 1642: 244-254, 2016 07 01.
Article in English | MEDLINE | ID: mdl-27026654

ABSTRACT

Protein tyrosine phosphate δ (PTPδ), one of the receptor type IIa protein tyrosine phosphates, is known for its roles in axon guidance, synapse formation, cell adhesion, and tumor suppression. Alternative splicing of this gene generates at least four (A-D) isoforms; however, the major isoform in vivo is yet to be determined. The protein localization has neither been revealed. We have generated anti-mouse PTPδ-specific monoclonal antibody and analyzed the protein expression in wild-type and Ptpδ knockout mice. Immunoblot analysis of various organs revealed that neuronal tissues express both C-and D-isoforms of PTPδ, whereas non-neuronal tissues express only C-isoform. Immunohistochemistry of wild-type or Ptpδ heterozygous sections showed that olfactory bulb, cerebral cortex, hippocampus, cerebellum, and several nuclei in brain stem exhibit moderate to strong positive signals. These signals were absent in Ptpδ knockout specimens. Higher magnification revealed differences between expression patterns of PTPδ mRNA and its protein product. In hippocampus, weak mRNA expression in CA1 stratum pyramidale but strong immunostaining in the stratum lacunosum moleculare was observed, suggesting the axonal expression of PTPδ in the entorhinal cortical afferents. Olfactory mitral cells exhibited mRNA expression in cell bodies and protein localization in their dendritic fields, glomerular and external plexiform layers. Nissl staining showed that the external plexiform layer was reduced in Ptpδ knockout mice. Golgi-impregnation confirmed the poor dendritic growth of homozygous mitral cells. These results suggest that PTPδ may localize in axons as well as in dendrites to regulate their elaboration in the central nervous system.


Subject(s)
Brain/enzymology , Neurons/enzymology , Receptor-Like Protein Tyrosine Phosphatases, Class 2/metabolism , Animals , Antibodies, Monoclonal/administration & dosage , Brain/immunology , Dendrites/enzymology , Isoenzymes/metabolism , Male , Mice , Mice, Knockout , Olfactory Bulb/cytology , Olfactory Bulb/enzymology , RNA, Messenger/metabolism , Rats , Rats, Wistar , Receptor-Like Protein Tyrosine Phosphatases, Class 2/genetics , Receptor-Like Protein Tyrosine Phosphatases, Class 2/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...