Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Org Lett ; 25(28): 5345-5349, 2023 07 21.
Article in English | MEDLINE | ID: mdl-37439572

ABSTRACT

Guanacastane diterpenoids with an unusual 5/7/6 tricyclic skeleton mainly produced by basidiomycete fungi represent a structurally intriguing class of natural products. While the chemical synthesis of several members has been achieved, the biochemical and genetic basis of their biosynthesis remain unknown. Herein, we present the identification and characterization of two crucial enzymes in the biosynthesis of guanacastane diterpenoids in Psathyrella candolleana. Heterologous expression reveals that PsaD, a typical class I diterpene synthase, catalyzes the cyclization of geranylgeranyl diphosphate to form a new guanacastane-type diterpene, guanacasta-1,3-diene (7). Moreover, we demonstrate that PsaA, a cytochrome P450 monooxygenase, can catalyze multiple oxidations of 7 to yield guanacastepene U (8). These results provide new opportunities for genome mining and metabolic engineering of guanacastane diterpenoids.


Subject(s)
Basidiomycota , Diterpenes , Basidiomycota/genetics , Diterpenes/chemistry
2.
Nano Lett ; 19(11): 8002-8009, 2019 11 13.
Article in English | MEDLINE | ID: mdl-31626554

ABSTRACT

Oncolytic adenovirus (OA) is an ideal candidate for clinical anticancer treatment, because it can specifically replicate in tumor cells with high titer. However, its systemic administration is still hindered, because of severely compromised antitumor efficacy. Herein, an engineered OA was innovatively developed by enwrapping OA with calcium and manganese carbonates (MnCaCs) biomineral shell, which could protect the virus from removal of the host immune system and prolong its in vivo circulation. Upon accumulating in tumor sites, MnCaCs readily dissolved under the acidic microenvironment, releasing Mn2+ that could convert endogenous H2O2 into oxygen (O2) and then enhance the duplication ability of OA, thus significantly increased the antitumor efficacy. Meanwhile, Mn2+ and the increased O2 individually endowed the T1 modal magnetic resonance imaging (MRI) and photoacoustic imaging (PAI) feasibility, providing real-time monitoring information for the therapy. This versatile engineered OA demonstrated its promise for visible and efficient oncolytic virotherapy by systemic administration.


Subject(s)
Adenoviridae/chemistry , Calcium Carbonate/chemistry , Carbonates/chemistry , Manganese/chemistry , Neoplasms/therapy , Oncolytic Virotherapy/methods , Oncolytic Viruses/chemistry , Adenoviridae/genetics , Animals , Genetic Engineering , Magnetic Resonance Imaging , Mice, Inbred BALB C , Neoplasms/diagnostic imaging , Oncolytic Viruses/genetics , Photoacoustic Techniques , Tumor Microenvironment
3.
J Neurochem ; 147(1): 40-57, 2018 10.
Article in English | MEDLINE | ID: mdl-29920676

ABSTRACT

Neuritin is a neurotrophic factor that is activated by neural activity and neurotrophins. Its major function is to promote neurite growth and branching; however, the underlying mechanisms are not fully understood. To address this issue, this study investigated the effects of neuritin on neurite and spine growth and intracellular Ca2+ concentration in rat cerebellar granule neurons (CGNs). Incubation of CGNs for 24 h with neuritin increased neurite length and spine density; this effect was mimicked by insulin and abolished by inhibiting insulin receptor (IR) or mitogen-activated protein kinase kinase/extracellular signal-regulated kinase (ERK) activity. Calcium imaging and western blot analysis revealed that neuritin enhanced the increase in intracellular Ca2+ level induced by high K+ , and stimulated the cell surface expression of CaV 1.2 and CaV 1.3 α subunits of the L-type calcium channel, which was suppressed by inhibition of IR or mitogen-activated protein kinase kinase/ERK. Treatment with inhibitors of L-type calcium channels, calmodulin, and calcineurin (CaN) abrogated the effects of neuritin on neurite length and spine density. A similar result was obtained by silencing nuclear factor of activated T cells c4, which is known to be activated by neuritin in CGNs. These results indicate that IR and ERK signaling as well as the Ca2+ /CaN/nuclear factor of activated T cells c4 axis mediate the effects of neuritin on neurite and spine growth in CGNs. OPEN PRACTICES: Open Science: This manuscript was awarded with the Open Materials Badge. For more information see: https://cos.io/our-services/open-science-badges/ Cover Image for this issue: doi: 10.1111/jnc.14195.


Subject(s)
Calcium Channels, L-Type/drug effects , Calcium Signaling/drug effects , Cerebellum/cytology , Dendritic Spines/drug effects , Neurites/drug effects , Neuropeptides/pharmacology , Animals , Calcium Channels/metabolism , Calcium Channels, L-Type/metabolism , Cerebellum/drug effects , Cerebellum/growth & development , Cytoplasmic Granules/drug effects , Female , GPI-Linked Proteins/pharmacology , Gene Silencing , Humans , Insulin/pharmacology , MAP Kinase Signaling System/drug effects , NFATC Transcription Factors/antagonists & inhibitors , NFATC Transcription Factors/genetics , Rats , Rats, Sprague-Dawley , Receptor, Insulin/antagonists & inhibitors
4.
Neurosci Bull ; 34(5): 759-768, 2018 Oct.
Article in English | MEDLINE | ID: mdl-29671217

ABSTRACT

Cyproheptadine (CPH), a first-generation antihistamine, enhances the delayed rectifier outward K+ current (IK) in mouse cortical neurons through a sigma-1 receptor-mediated protein kinase A pathway. In this study, we aimed to determine the effects of CPH on neuronal excitability in current-clamped pyramidal neurons in mouse medial prefrontal cortex slices. CPH (10 µmol/L) significantly reduced the current density required to generate action potentials (APs) and increased the instantaneous frequency evoked by a depolarizing current. CPH also depolarized the resting membrane potential (RMP), decreased the delay time to elicit an AP, and reduced the spike threshold potential. This effect of CPH was mimicked by a sigma-1 receptor agonist and eliminated by an antagonist. Application of tetraethylammonium (TEA) to block IK channels hyperpolarized the RMP and reduced the instantaneous frequency of APs. TEA eliminated the effects of CPH on AP frequency and delay time, but had no effect on spike threshold or RMP. The current-voltage relationship showed that CPH increased the membrane depolarization in response to positive current pulses and hyperpolarization in response to negative current pulses, suggesting that other types of membrane ion channels might also be affected by CPH. These results suggest that CPH increases the excitability of medial prefrontal cortex neurons by regulating TEA-sensitive IK channels as well as other TEA-insensitive K+ channels, probably ID and inward-rectifier Kir channels. This effect of CPH may explain its apparent clinical efficacy as an antidepressant and antipsychotic.


Subject(s)
Cyproheptadine/pharmacology , Histamine H1 Antagonists/pharmacology , Prefrontal Cortex/drug effects , Pyramidal Cells/drug effects , Animals , Female , Membrane Potentials/drug effects , Membrane Potentials/physiology , Mice, Inbred C57BL , Patch-Clamp Techniques , Potassium Channel Blockers/pharmacology , Potassium Channels/metabolism , Prefrontal Cortex/physiology , Pyramidal Cells/physiology , Receptors, sigma/agonists , Receptors, sigma/metabolism , Tetraethylammonium/pharmacology , Tissue Culture Techniques
5.
Acta Pharmacol Sin ; 39(9): 1414-1420, 2018 Sep.
Article in English | MEDLINE | ID: mdl-29595190

ABSTRACT

Neuritin is a member of the neurotrophic factor family, which is activated by neural activity and neurotrophins, and promotes neurite growth and branching. It has shown to play an important role in neuronal plasticity and regeneration. It is also involved in other biological processes such as angiogenesis, tumorigenesis and immunomodulation. Thus far, however, the primary mechanisms of neuritin, including whether or not it acts through a receptor or which downstream signals might be activated following binding, are not fully understood. Recent evidence suggests that neuritin may be a potential therapeutic target in several neurodegenerative diseases. This review focuses on the recent advances in studies regarding the newly identified functions of neuritin and the signaling pathways related to these functions. We also discuss current hot topics and difficulties in neuritin research.


Subject(s)
Neuropeptides/physiology , Signal Transduction/physiology , Animals , GPI-Linked Proteins/physiology , Humans , Mental Disorders/etiology , Mental Disorders/physiopathology , Neurogenesis/physiology , Neuronal Plasticity/physiology , Synapses/physiology
6.
Sci Rep ; 7: 44521, 2017 03 17.
Article in English | MEDLINE | ID: mdl-28303965

ABSTRACT

Mounting evidence suggests that exposure to radiofrequency electromagnetic radiation (RF-EMR) can influence learning and memory in rodents. In this study, we examined the effects of single exposure to 1.8 GHz RF-EMR for 30 min on subsequent recognition memory in mice, using the novel object recognition task (NORT). RF-EMR exposure at an intensity of >2.2 W/kg specific absorption rate (SAR) power density induced a significant density-dependent increase in NORT index with no corresponding changes in spontaneous locomotor activity. RF-EMR exposure increased dendritic-spine density and length in hippocampal and prefrontal cortical neurons, as shown by Golgi staining. Whole-cell recordings in acute hippocampal and medial prefrontal cortical slices showed that RF-EMR exposure significantly altered the resting membrane potential and action potential frequency, and reduced the action potential half-width, threshold, and onset delay in pyramidal neurons. These results demonstrate that exposure to 1.8 GHz RF-EMR for 30 min can significantly increase recognition memory in mice, and can change dendritic-spine morphology and neuronal excitability in the hippocampus and prefrontal cortex. The SAR in this study (3.3 W/kg) was outside the range encountered in normal daily life, and its relevance as a potential therapeutic approach for disorders associated with recognition memory deficits remains to be clarified.


Subject(s)
Electromagnetic Fields/adverse effects , Electromagnetic Radiation , Pattern Recognition, Visual/radiation effects , Pyramidal Cells/radiation effects , Action Potentials/radiation effects , Animals , Dendritic Spines/pathology , Dendritic Spines/radiation effects , Hippocampus/physiopathology , Hippocampus/radiation effects , Memory , Memory Disorders/etiology , Memory Disorders/physiopathology , Mice , Pyramidal Cells/pathology , Radio Waves/adverse effects
7.
Sci Rep ; 6: 28653, 2016 06 29.
Article in English | MEDLINE | ID: mdl-27353765

ABSTRACT

Growth differentiation factor-15 (GDF-15) has been implicated in ischemic brain injury and synapse development, but its involvement in modulating neuronal excitability and synaptic transmission remain poorly understood. In this study, we investigated the effects of GDF-15 on non-evoked miniature excitatory post-synaptic currents (mEPSCs) and neurotransmitter release in the medial prefrontal cortex (mPFC) in mice. Incubation of mPFC slices with GDF-15 for 60 min significantly increased the frequency of mEPSCs without effect on their amplitude. GDF-15 also significantly elevated presynaptic glutamate release, as shown by HPLC. These effects were blocked by dual TGF-ß type I receptor (TßRI) and TGF-ß type II receptor (TßRII) antagonists, but not by a TßRI antagonist alone. Meanwhile, GDF-15 enhanced pERK level, and inhibition of MAPK/ERK activity attenuated the GDF-15-induced increases in mEPSC and glutamate release. Blocking T-type calcium channels reduced the GDF-15 induced up-regulation of synaptic transmission. Membrane-protein extraction and use of an intracellular protein-transport inhibitor showed that GDF-15 promoted CaV3.1 and CaV3.3 α-subunit expression by trafficking to the membrane. These results confirm previous findings in cerebellar granule neurons, in which GDF-15 induces its neurobiological effects via TßRII and activation of the ERK pathway, providing novel insights into the mechanism of GDF-15 function in cortical neurons.


Subject(s)
Calcium Channels, T-Type/biosynthesis , Glutamic Acid/metabolism , Growth Differentiation Factor 15/metabolism , MAP Kinase Signaling System/physiology , Prefrontal Cortex/metabolism , Synaptic Transmission/physiology , Animals , Female , Mice , Protein Serine-Threonine Kinases/metabolism , Receptor, Transforming Growth Factor-beta Type II , Receptors, Transforming Growth Factor beta/metabolism
8.
J Biol Chem ; 291(33): 17369-81, 2016 08 12.
Article in English | MEDLINE | ID: mdl-27307045

ABSTRACT

Neuritin is an important neurotrophin that regulates neural development, synaptic plasticity, and neuronal survival. Elucidating the downstream molecular signaling is important for potential therapeutic applications of neuritin in neuronal dysfunctions. We previously showed that neuritin up-regulates transient potassium outward current (IA) subunit Kv4.2 expression and increases IA densities, in part by activating the insulin receptor signaling pathway. Molecular mechanisms of neuritin-induced Kv4.2 expression remain elusive. Here, we report that the Ca(2+)/calcineurin (CaN)/nuclear factor of activated T-cells (NFAT) c4 axis is required for neuritin-induced Kv4.2 transcriptional expression and potentiation of IA densities in cerebellum granule neurons. We found that neuritin elevates intracellular Ca(2+) and increases Kv4.2 expression and IA densities; this effect was sensitive to CaN inhibition and was eliminated in Nfatc4(-/-) mice but not in Nfatc2(-/-) mice. Stimulation with neuritin significantly increased nuclear accumulation of NFATc4 in cerebellum granule cells and HeLa cells, which expressed IR. Furthermore, NFATc4 was recruited to the Kv4.2 gene promoter loci detected by luciferase reporter and chromatin immunoprecipitation assays. More importantly, data obtained from cortical neurons following adeno-associated virus-mediated overexpression of neuritin indicated that reduced neuronal excitability and increased formation of dendritic spines were abrogated in the Nfatc4(-/-) mice. Together, these data demonstrate an indispensable role for the CaN/NFATc4 signaling pathway in neuritin-regulated neuronal functions.


Subject(s)
Calcineurin/metabolism , Calcium Signaling/physiology , Calcium/metabolism , Dendritic Spines/metabolism , Gene Expression Regulation/physiology , NFATC Transcription Factors/metabolism , Nerve Tissue Proteins/metabolism , Neuropeptides/metabolism , Shal Potassium Channels/biosynthesis , Animals , Calcineurin/genetics , Cerebellum/metabolism , Dendritic Spines/genetics , GPI-Linked Proteins/genetics , GPI-Linked Proteins/metabolism , HeLa Cells , Humans , Mice , Mice, Knockout , NFATC Transcription Factors/genetics , Nerve Tissue Proteins/genetics , Neuropeptides/genetics , Shal Potassium Channels/genetics
9.
Sci Rep ; 5: 11768, 2015 Jul 03.
Article in English | MEDLINE | ID: mdl-26138388

ABSTRACT

Animal studies have shown that electromagnetic field exposure may interfere with the activity of brain cells, thereby generating behavioral and cognitive disturbances. However, the underlying mechanisms and possible preventions are still unknown. In this study, we used a mouse model to examine the effects of exposure to extremely low-frequency (50 Hz) electromagnetic fields (ELF MFs) on a recognition memory task and morphological changes of hippocampal neurons. The data showed that ELF MFs exposure (1 mT, 12 h/day) induced a time-dependent deficit in novel object associative recognition memory and also decreased hippocampal dendritic spine density. This effect was observed without corresponding changes in spontaneous locomotor activity and was transient, which has only been seen after exposing mice to ELF MFs for 7-10 days. The over-expression of hippocampal neuritin, an activity-dependent neurotrophic factor, using an adeno-associated virus (AAV) vector significantly increased the neuritin level and dendritic spine density. This increase was paralleled with ELF MFs exposure-induced deficits in recognition memory and reductions of dendritic spine density. Collectively, our study provides evidence for the association between ELF MFs exposure, impairment of recognition memory, and resulting changes in hippocampal dendritic spine density. Neuritin prevented this ELF MFs-exposure-induced effect by increasing the hippocampal spine density.


Subject(s)
Electromagnetic Fields/adverse effects , Hippocampus/physiopathology , Memory Disorders/prevention & control , Nerve Tissue Proteins/physiology , Animals , Dendritic Spines/pathology , Dependovirus/genetics , Female , GPI-Linked Proteins/physiology , Genetic Vectors , Hippocampus/metabolism , Hippocampus/pathology , Memory Disorders/etiology , Memory Disorders/physiopathology , Mice, Inbred ICR , Pattern Recognition, Visual , Protective Factors , Recognition, Psychology
10.
J Cell Mol Med ; 18(6): 1060-70, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24548607

ABSTRACT

Although melatonin (MT) has been reported to protect cells against oxidative damage induced by electromagnetic radiation, few reports have addressed whether there are other protective mechanisms. Here, we investigated the effects of MT on extremely low-frequency electromagnetic field (ELF-EMF)-induced Nav activity in rat cerebellar granule cells (GCs). Exposing cerebellar GCs to ELF-EMF for 60 min. significantly increased the Nav current (INa ) densities by 62.5%. MT (5 µM) inhibited the ELF-EMF-induced INa increase. This inhibitory effect of MT is mimicked by an MT2 receptor agonist and was eliminated by an MT2 receptor antagonist. The Nav channel steady-state activation curve was significantly shifted towards hyperpolarization by ELF-EMF stimulation but remained unchanged by MT in cerebellar GC that were either exposed or not exposed to ELF-EMF. ELF-EMF exposure significantly increased the intracellular levels of phosphorylated PKA in cerebellar GCs, and both MT and IIK-7 did not reduce the ELF-EMF-induced increase in phosphorylated PKA. The inhibitory effects of MT on ELF-EMF-induced Nav activity was greatly reduced by the calmodulin inhibitor KN93. Calcium imaging showed that MT did not increase the basal intracellular Ca(2+) level, but it significantly elevated the intracellular Ca(2+) level evoked by the high K(+) stimulation in cerebellar GC that were either exposed or not exposed to ELF-EMF. In the presence of ruthenium red, a ryanodine-sensitive receptor blocker, the MT-induced increase in intracellular calcium levels was reduced. Our data show for the first time that MT protects against neuronal INa that result from ELF-EMF exposure through Ca(2+) influx-induced Ca(2+) release.


Subject(s)
Calcium/metabolism , Cerebellum/cytology , Cytoplasmic Granules/metabolism , Electromagnetic Fields/adverse effects , Melatonin/pharmacology , Protective Agents/pharmacology , Voltage-Gated Sodium Channels/metabolism , Animals , Antioxidants/pharmacology , Cells, Cultured , Cerebellum/drug effects , Cerebellum/metabolism , Cerebellum/radiation effects , Cytoplasmic Granules/drug effects , Cytoplasmic Granules/radiation effects , Male , Mice , Oxidation-Reduction , Patch-Clamp Techniques , Phosphorylation/drug effects , Phosphorylation/radiation effects , Rats , Rats, Sprague-Dawley , Signal Transduction/drug effects , Signal Transduction/radiation effects
11.
Am J Physiol Cell Physiol ; 305(2): C197-206, 2013 Jul 15.
Article in English | MEDLINE | ID: mdl-23703525

ABSTRACT

Neuregulin-1 (NRG-1) is a member of a family of neurotrophic factors that is required for the differentiation, migration, and development of neurons. NRG-1 signaling is thought to contribute to both neuronal development and the neuropathology of schizophrenia, which is believed to be a neurodevelopmental disorder. However, few studies have investigated the role of NRG-1 on voltage-gated ion channels. In this study, we report that NRG-1 specifically increases the density of transient outward K(+) currents (IA) in rat cerebellar granule neurons (CGNs) in a time-dependent manner without modifying the activation or inactivation properties of IA channels. The increase in IA density is mediated by increased protein expression of Kv4.2, the main α-subunit of the IA channel, most likely by upregulation of translation. The effect of NRG-1 on IA density and Kv4.2 expression was only significant in immature neurons. Mechanistically, both Akt and mammalian target of rapamycin (mTOR) signaling pathways are required for the increased NRG-1-induced IA density and expression of Kv4.2. Moreover, pharmacological blockade of the ErbB4 receptor reduced the effect of NRG-1 on IA density and Kv4.2 induction. Our data reveal, for the first time, that stimulation of ErbB4 signaling by NRG-1 upregulates the expression of K(+) channel proteins via activation of the Akt/mTOR signaling pathway and plays an important role in neuronal development and maturation. NRG1 does not acutely change IA and delayed-rectifier outward (IK) of rat CGNs, suggesting that it may not alter excitability of immature neurons by altering potassium channel property.


Subject(s)
ErbB Receptors/metabolism , Neuregulin-1/metabolism , Potassium/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Shal Potassium Channels/metabolism , TOR Serine-Threonine Kinases/metabolism , Animals , Cell Movement , ErbB Receptors/genetics , Gene Expression Regulation/physiology , Membrane Potentials , Neuregulin-1/genetics , Proto-Oncogene Proteins c-akt/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Receptor, ErbB-4 , Shal Potassium Channels/genetics , Signal Transduction , TOR Serine-Threonine Kinases/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...