Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 23
1.
Ageing Res Rev ; 96: 102252, 2024 Apr.
Article En | MEDLINE | ID: mdl-38442748

Chronic cerebral hypoperfusion (CCH) is a common mechanism of acute brain injury due to impairment of blood flow to the brain. Moreover, a prolonged lack of oxygen supply may result in cerebral infarction or global ischemia, which subsequently causes long-term memory impairment. Research on using Clitoria ternatea root extract for treating long-term memory has been studied extensively. However, the bioactive compound contributing to its neuroprotective effects remains uncertain. In the present study, we investigate the effects of clitorienolactone A (CLA) and B (CLB) from the roots of Clitoria ternatea extract on hippocampal neuroplasticity in rats induced by CCH. CLA and CLB were obtained using column chromatography. The rat model of CCH was induced using two-vessel occlusion surgery (2VO). The 2VO rats were given 10 mg/kg of CLA and CLB orally, followed by hippocampal neuroplasticity recording using in vivo electrophysiological. Rats received CLA and CLB (10 mg/kg) significantly reversed the impairment of long-term potentiation following 2VO surgery. Furthermore, we investigate the effect of CLA and CLB on the calcium channel using the calcium imaging technique. During hypoxia, CLA and CLB sustain the increase in intracellular calcium levels. We next predict the binding interactions of CLA and CLB against NMDA receptors containing GluN2A and GluN2B subunits using in silico molecular docking. Our result found that both CLA and CLB exhibited lower binding affinity against GluN2A and GluN2B subunits. Our findings demonstrated that bioactive compounds from Clitoria ternatea improved long-term memory deficits in the chronic cerebral hypoperfusion rat model via calcium uptake. Hence, CLA and CLB could be potential therapeutic tools for treating cognitive dysfunction.


Brain Ischemia , Clitoria , Rats , Humans , Animals , Clitoria/chemistry , Calcium Channels/pharmacology , Calcium Channels/therapeutic use , Long-Term Potentiation , Calcium , Molecular Docking Simulation , Brain Ischemia/drug therapy , Hippocampus , Maze Learning/physiology
2.
J Drug Target ; 32(4): 413-422, 2024 Apr.
Article En | MEDLINE | ID: mdl-38345028

BACKGROUND: Blocking Transient Receptor Potential Melastatin 4 (TRPM4) in rodents by our antibody M4P has shown to attenuate cerebral ischaemia-reperfusion injury. Since M4P does not interact with human TRPM4, the therapeutic potential of blocking human TRPM4 remains unclear. We developed a monoclonal antibody M4M that inhibited human TRPM4 in cultured cells. However, M4M has no effect on stroke outcome in wild-type rats. Therefore, M4M needs to be evaluated on animal models expressing human TRPM4. METHODS: We generated a humanised rat model using the CRISPR/Cas technique to knock-in (KI) the human TRPM4 antigen sequence. RESULTS: In primary neurons from human TRPM4 KI rats, M4M binds to hypoxic neurons, but not normoxic nor wild-type neurons. Electrophysiological studies showed that M4M blocked ATP depletion-induced activation of TRPM4 and inhibited hypoxia-associated cell volume increase. In a stroke model, administration of M4M reduced infarct volume in KI rats. Rotarod test and Neurological deficit score revealed improvement following M4M treatment. CONCLUSION: M4M selectively binds and inhibits hypoxia-induced human TRPM4 channel activation in neurons from the humanised rat model, with no effect on healthy neurons. Use of M4M in stroke rats showed functional improvements, suggesting the potential for anti-human TRPM4 antibodies in treating acute ischaemic stroke patients.


Brain Ischemia , Reperfusion Injury , Stroke , TRPM Cation Channels , Transient Receptor Potential Channels , Rats , Humans , Animals , Stroke/drug therapy , Transient Receptor Potential Channels/therapeutic use , Antibodies, Monoclonal/pharmacology , Brain Ischemia/drug therapy , TRPM Cation Channels/metabolism , Reperfusion Injury/drug therapy , Hypoxia
3.
Neurobiol Dis ; 191: 106408, 2024 Feb.
Article En | MEDLINE | ID: mdl-38199274

Excitotoxicity arises from unusually excessive activation of excitatory amino acid receptors such as glutamate receptors. Following an energy crisis, excitotoxicity is a major cause for neuronal death in neurological disorders. Many glutamate antagonists have been examined for their efficacy in mitigating excitotoxicity, but failed to generate beneficial outcome due to their side effects on healthy neurons where glutamate receptors are also blocked. In this study, we found that during chronic hypoxia there is upregulation and activation of a nonselective cation channel TRPM4 that contributes to the depolarized neuronal membrane potential and enhanced glutamate-induced calcium entry. TRPM4 is involved in modulating neuronal membrane excitability and calcium signaling, with a complex and multifaceted role in the brain. Here, we inhibited TRPM4 using a newly developed blocking antibody M4P, which could repolarize the resting membrane potential and ameliorate calcium influx upon glutamate stimulation. Importantly, M4P did not affect the functions of healthy neurons as the activity of TRPM4 channel is not upregulated under normoxia. Using a rat model of chronic hypoxia with both common carotid arteries occluded, we found that M4P treatment could reduce apoptosis in the neurons within the hippocampus, attenuate long-term potentiation impairment and improve the functions of learning and memory in this rat model. With specificity to hypoxic neurons, TRPM4 blocking antibody can be a novel way of controlling excitotoxicity with minimal side effects that are common among direct blockers of glutamate receptors.


Glutamic Acid , TRPM Cation Channels , Rats , Animals , Glutamic Acid/metabolism , Calcium/metabolism , Receptors, Glutamate/metabolism , Neurons/metabolism , Hypoxia/metabolism , TRPM Cation Channels/metabolism
4.
Mol Neurobiol ; 60(10): 5931-5943, 2023 Oct.
Article En | MEDLINE | ID: mdl-37380823

Neuronal swelling is a pathological feature of stroke which contributes to the formation of cytotoxic edema. Under hypoxic condition, aberrant accumulation of sodium and chloride ions inside neurons increases osmotic pressure, leading to cell volume increase. Sodium entry pathway in neurons has been studied extensively. Here, we determine whether SLC26A11 is the major chloride entry pathway under hypoxia and could be the target for protection against ischemic stroke. In this study, electrophysiological properties of chloride current in primary cultured neurons were characterized using low chloride solution, 4,4'-diisothiocyano-2,2'-stilbenedisulfonic acid, and SLC26A11-specific siRNA under physiological conditions or ATP-depleted conditions. In vivo effect of SLC26A11 was evaluated on a rat stroke reperfusion model. We found that SLC26A11 mRNA in primary cultured neurons was upregulated as early as 6 h after oxygen glucose deprivation, and later, the protein level was elevated accordingly. Blockade of SLC26A11 activity could reduce chloride entry and attenuate hypoxia-induced neuronal swelling. In the animal stroke model, SLC26A11 upregulation was mainly located in surviving neurons close to the infarct core. SLC26A11 inhibition ameliorates infarct formation and improves functional recovery. These findings demonstrate that SLC26A11 is a major pathway for chloride entry in stroke, contributing to neuronal swelling. Inhibition of SLC26A11 could be a novel therapeutic strategy for stroke.


Brain Ischemia , Reperfusion Injury , Stroke , Rats , Animals , Chlorides , Stroke/complications , Stroke/drug therapy , Stroke/metabolism , Hypoxia/pathology , Edema , Reperfusion Injury/complications , Reperfusion Injury/drug therapy , Infarction , Sodium/metabolism , Glucose , Brain Ischemia/pathology
5.
Biomedicines ; 11(5)2023 May 19.
Article En | MEDLINE | ID: mdl-37239151

Reperfusion therapy for acute ischemic stroke aims to restore the blood flow of occluded blood vessels. However, successful recanalization is often associated with disruption of the blood-brain barrier, leading to reperfusion injury. Delayed recanalization increases the risk of severe reperfusion injury, including severe cerebral edema and hemorrhagic transformation. The TRPM4-blocking antibody M4P has been shown to alleviate reperfusion injury and improve functional outcomes in animal models of early stroke reperfusion. In this study, we examined the role of M4P in a clinically relevant rat model of delayed stroke reperfusion in which the left middle cerebral artery was occluded for 7 h. To mimic the clinical scenario, M4P or control IgG was administered 1 h before recanalization. Immunostaining showed that M4P treatment improved vascular morphology after stroke. Evans blue extravasation demonstrated attenuated vascular leakage following M4P treatment. With better vascular integrity, cerebral perfusion was improved, leading to a reduction of infarct volume and animal mortality rate. Functional outcome was evaluated by the Rotarod test. As more animals with severe injuries died during the test in the control IgG group, we observed no difference in functional outcomes in the surviving animals. In conclusion, we identified the potential of TRPM4 blocking antibody M4P to ameliorate vascular injury during delayed stroke reperfusion. If combined with reperfusion therapy, M4P has the potential to improve current stroke management.

6.
Sci Rep ; 12(1): 19562, 2022 11 15.
Article En | MEDLINE | ID: mdl-36380063

Mouse monoclonal antibody M4M was recently designed to block human TRPM4 channel. The polypeptide for generating M4M is composed of peptide A1 between the transmembrane segment 5 (S5) and the pore, and a second peptide A2 between the pore and the transmembrane segment 6 (S6). Using peptide microarray, a 4-amino acid sequence EPGF within the A2 was identified to be the binding epitope for M4M. Substitution of EPGF with other amino acids greatly reduced binding affinity. Structural analysis of human TRPM4 structure indicates that EPGF is located externally to the channel pore. A1 is close to the EPGF binding epitope in space, albeit separated by a 37-amino acid peptide. Electrophysiological study reveals that M4M could block human TRPM4, but with no effect on rodent TRPM4 which shares a different amino acid sequence ERGS for the binding motif. Our results demonstrate that M4M is a specific inhibitor for human TRPM4.


Antibodies, Monoclonal , TRPM Cation Channels , Mice , Animals , Humans , Epitopes , Antibodies, Monoclonal/metabolism , Amino Acid Sequence , Peptides/metabolism , Mutation, Missense , TRPM Cation Channels/genetics , TRPM Cation Channels/metabolism
7.
Sci Rep ; 11(1): 10411, 2021 05 17.
Article En | MEDLINE | ID: mdl-34002002

TRPM4 is a calcium-activated non-selective monovalent cation channel implicated in diseases such as stroke. Lack of potent and selective inhibitors remains a major challenge for studying TRPM4. Using a polypeptide from rat TRPM4, we have generated a polyclonal antibody M4P which could alleviate reperfusion injury in a rat model of stroke. Here, we aim to develop a monoclonal antibody that could block human TRPM4 channel. Two mouse monoclonal antibodies M4M and M4M1 were developed to target an extracellular epitope of human TRPM4. Immunohistochemistry and western blot were used to characterize the binding of these antibodies to human TRPM4. Potency of inhibition was compared using electrophysiological methods. We further evaluated the therapeutic potential on a rat model of middle cerebral artery occlusion. Both M4M and M4M1 could bind to human TRPM4 channel on the surface of live cells. Prolonged incubation with TRPM4 blocking antibody internalized surface TRPM4. Comparing to M4M1, M4M is more effective in blocking human TRPM4 channel. In human brain microvascular endothelial cells, M4M successfully inhibited TRPM4 current and ameliorated hypoxia-induced cell swelling. Using wild type rats, neither antibody demonstrated therapeutic potential on stroke. Human TRPM4 channel can be blocked by a monoclonal antibody M4M targeting a key antigenic sequence. For future clinical translation, the antibody needs to be humanized and a transgenic animal carrying human TRPM4 sequence is required for in vivo characterizing its therapeutic potential.


Antibodies, Monoclonal/pharmacology , Infarction, Middle Cerebral Artery/drug therapy , Reperfusion Injury/prevention & control , TRPM Cation Channels/antagonists & inhibitors , Animals , Antibodies, Monoclonal/therapeutic use , Brain/cytology , Brain/drug effects , Brain/pathology , Disease Models, Animal , Drug Evaluation, Preclinical , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Endothelial Cells/pathology , HEK293 Cells , Humans , Infarction, Middle Cerebral Artery/complications , Male , Patch-Clamp Techniques , Rats , Reperfusion Injury/etiology , Reperfusion Injury/pathology , TRPM Cation Channels/metabolism
8.
Front Cell Dev Biol ; 8: 562584, 2020.
Article En | MEDLINE | ID: mdl-33195194

In stroke and other neurological diseases, Transient Receptor Potential Melastatin 4 (TRPM4) has been reported to cause oncotic cell death which is due to an excessive influx of sodium ions. Following stroke, hypoxia condition activates TRPM4 channel, and the sodium influx via TRPM4 is further enhanced by an increased TRPM4 expression. However, the effect of TRPM4 inhibition on oncotic cell death, particularly during the acute stage, remains largely unknown. Recently, we have developed a polyclonal antibody M4P that specifically inhibits TRPM4 channel. M4P blocks the channel via binding to a region close to the channel pore from extracellular space. Using M4P, we evaluated the acute effect of blocking TRPM4 in neurons, astrocytes, and vascular endothelial cells. In a rat stroke model, M4P co-localized with neuronal marker NeuN and endothelial marker vWF, whereas few GFAP positive astrocytes were stained by M4P in the ipsilateral hemisphere. When ATP was acutely depleted in cultured cortical neurons and microvascular endothelial cells, cell swelling was induced. Application of M4P significantly blocked TRPM4 current and attenuated oncosis. TUNEL assay, PI staining and western blot on cleaved Caspase-3 revealed that M4P could ameliorate apoptosis after 24 h hypoxia exposure. In contrast, acute ATP depletion in cultured astrocytes failed to demonstrate an increase of cell volume, and application of M4P or control IgG had no effect on cell volume change. When TRPM4 was overexpressed in astrocytes, acute ATP depletion successfully induced oncosis which could be suppressed by M4P treatment. Our results demonstrate that comparing to astrocytes, neurons, and vascular endothelial cells are more vulnerable to hypoxic injury. During the acute stage of stroke, blocking TRPM4 channel could protect neurons and vascular endothelial cells from oncotic cell death.

9.
Pflugers Arch ; 471(11-12): 1455-1466, 2019 12.
Article En | MEDLINE | ID: mdl-31664513

Reperfusion therapy is currently the gold standard treatment for acute ischemic stroke. However, reperfusion injuries such as oedema and haemorrhagic transformation largely limit the use of this potent treatment to a narrow time window. Recently, transient receptor potential melastatin 4 (TRPM4) channel has emerged as a potential target for vascular protection in stroke management. Non-specificity and side effects are major concerns for current TRPM4 blockers. The present study was undertaken to develop a novel TRPM4 blocker for stroke management. We report the generation of a TRPM4-specific antibody M4P which binds to a region close to the channel pore. M4P could inhibit TRPM4 current and downregulate TRPM4 surface expression, therefore prevent hypoxia-induced cell swelling. In the rat model of 3-h stroke reperfusion, application of M4P at 2 h after occlusion ameliorated reperfusion injury by improving blood-brain barrier integrity, and enhanced functional recovery. Our results demonstrate that TRPM4 blockade could attenuate reperfusion injury in stroke recanalization. When applied together with reperfusion treatments, TRPM4 blocking antibody has the potential to extend the therapeutic time window for acute ischemic stroke.


Antibodies, Monoclonal/pharmacology , Reperfusion Injury/drug therapy , Stroke/drug therapy , TRPM Cation Channels/antagonists & inhibitors , Animals , Blood-Brain Barrier/metabolism , Brain Ischemia/drug therapy , Brain Ischemia/metabolism , Disease Models, Animal , Female , Male , Rats , Rats, Wistar , Reperfusion Injury/metabolism , Stroke/metabolism , Up-Regulation/drug effects
10.
Biotechnol J ; 12(12)2017 Dec.
Article En | MEDLINE | ID: mdl-28762648

Neural crest stem cells (NCSCs) are a transient and multipotent cell population giving rise to various cell types with clinical importance. Isolation of human NCSCs is extremely challenging that limits our knowledge about neural crest development and application. Here, a defined protocol to efficiently direct human embryonic stem cells (hESCs) to NCSCs and multiple neural crest lineages is presented. A unique combination of small molecule inhibitors and growth factors is employed to generate NCSCs from hESCs through a neuroectoderm stage. The self-renewal and multipotent capacities of hESC-derived NCSCs are assessed subsequently. In the feeder-free system, hESC-derived NCSCs (P75+ /HNK1+ /AP2α+ /PAX6- ) in high purity are efficiently generated following neuroectodermal restriction. They can be propagated and differentiated toward multiple neural crest lineages in vitro, such as functional peripheral neurons (ß-tubulin III+ /peripherin+ ), mesenchymal stem cells (CD73+ CD90+ CD105+ ), and corneal keratocytes (keratocan+ ). The in vivo developmental potential of hESC-derived NCSCs is confirmed using zebrafish embryos. This report is the first demonstration of efficient differentiation of hESCs into corneal keratocytes as a monolayer in a feeder-free system. Considering the high efficacy of NCSC generation, this new method will be a useful tool for future clinical organ repair and regeneration, such as peripheral nerve regeneration and corneal repair.


Corneal Keratocytes/cytology , Human Embryonic Stem Cells/cytology , Neural Crest/cytology , Neural Stem Cells/cytology , Neurogenesis/physiology , Animals , Cell Culture Techniques/methods , Cells, Cultured , Embryo, Nonmammalian , Flow Cytometry , Humans , Neurons/cytology , Zebrafish
11.
Stem Cells Transl Med ; 6(9): 1803-1814, 2017 09.
Article En | MEDLINE | ID: mdl-28650520

Recent reports have indicated human embryonic stem cells-derived midbrain dopamine (mDA) neurons as proper cell resources for use in Parkinson's disease (PD) therapy. Nevertheless, no detailed and systematic study has been conducted to identify which differentiation stages of mDA cells are most suitable for transplantation in PD therapy. Here, we transplanted three types of mDA cells, DA progenitors (differentiated in vitro for 16 days [D16]), immature DA neurons (D25), and DA neurons (D35), into PD mice and found that all three types of cells showed high viability and strong neuronal differentiation in vivo. Both D25 and D35 cells showed neuronal maturation and differentiation toward TH+ cells and, accordingly, satisfactory behavioral functional recovery. However, transplanted D16 cells were less capable of producing functional recovery. These findings provide a valuable guideline for standardizing the differentiation stage of the transplantable cells used in clinical cell therapy for PD. Stem Cells Translational Medicine 2017;6:1803-1814.


Dopaminergic Neurons/cytology , Mesencephalon/cytology , Neural Stem Cells/cytology , Parkinson Disease/therapy , Stem Cell Transplantation/methods , Animals , Cells, Cultured , Dopaminergic Neurons/transplantation , Humans , Male , Mice , Mice, Inbred NOD , Mice, SCID , Neural Stem Cells/transplantation , Neurogenesis
12.
Proc Natl Acad Sci U S A ; 112(32): 9996-10001, 2015 Aug 11.
Article En | MEDLINE | ID: mdl-26216970

Glucose stimulates insulin secretion from ß-cells by increasing intracellular Ca(2+). Ca(2+) then binds to synaptotagmin-7 as a major Ca(2+) sensor for exocytosis, triggering secretory granule fusion and insulin secretion. In type-2 diabetes, insulin secretion is impaired; this impairment is ameliorated by glucagon-like peptide-1 (GLP-1) or by GLP-1 receptor agonists, which improve glucose homeostasis. However, the mechanism by which GLP-1 receptor agonists boost insulin secretion remains unclear. Here, we report that GLP-1 stimulates protein kinase A (PKA)-dependent phosphorylation of synaptotagmin-7 at serine-103, which enhances glucose- and Ca(2+)-stimulated insulin secretion and accounts for the improvement of glucose homeostasis by GLP-1. A phospho-mimetic synaptotagmin-7 mutant enhances Ca(2+)-triggered exocytosis, whereas a phospho-inactive synaptotagmin-7 mutant disrupts GLP-1 potentiation of insulin secretion. Our findings thus suggest that synaptotagmin-7 is directly activated by GLP-1 signaling and may serve as a drug target for boosting insulin secretion. Moreover, our data reveal, to our knowledge, the first physiological modulation of Ca(2+)-triggered exocytosis by direct phosphorylation of a synaptotagmin.


Glucagon-Like Peptide 1/metabolism , Insulin-Secreting Cells/metabolism , Insulin/metabolism , Synaptotagmins/metabolism , Amino Acid Sequence , Animals , Colforsin/pharmacology , Conserved Sequence , Cyclic AMP/pharmacology , Cyclic AMP-Dependent Protein Kinases/metabolism , Evolution, Molecular , Exenatide , Exocytosis/drug effects , Glucagon-Like Peptide-1 Receptor , Glucose/pharmacology , HEK293 Cells , Humans , Insulin Secretion , Insulin-Secreting Cells/drug effects , Mice, Knockout , Molecular Sequence Data , Mutation/genetics , Peptides/pharmacology , Phosphorylation/drug effects , Phosphoserine/metabolism , Rats , Receptors, Glucagon/metabolism , Synaptotagmins/chemistry , Venoms/pharmacology
13.
EMBO Rep ; 15(6): 714-22, 2014 Jun.
Article En | MEDLINE | ID: mdl-24711543

While molecular regulation of insulin granule exocytosis is relatively well understood, insulin granule biogenesis and maturation and its influence on glucose homeostasis are relatively unclear. Here, we identify a novel protein highly expressed in insulin-secreting cells and name it BIG3 due to its similarity to BIG/GBF of the Arf-GTP exchange factor (GEF) family. BIG3 is predominantly localized to insulin- and clathrin-positive trans-Golgi network (TGN) compartments. BIG3-deficient insulin-secreting cells display increased insulin content and granule number and elevated insulin secretion upon stimulation. Moreover, BIG3 deficiency results in faster processing of proinsulin to insulin and chromogranin A to ß-granin in ß-cells. BIG3-knockout mice exhibit postprandial hyperinsulinemia, hyperglycemia, impaired glucose tolerance, and insulin resistance. Collectively, these results demonstrate that BIG3 negatively modulates insulin granule biogenesis and insulin secretion and participates in the regulation of systemic glucose homeostasis.


Homeostasis/genetics , Insulin-Secreting Cells/metabolism , Insulin/metabolism , Proteins/metabolism , Secretory Vesicles/chemistry , Animals , Calorimetry, Indirect , Glucose/physiology , Homeostasis/physiology , Hyperglycemia/genetics , Insulin/analysis , Insulin Resistance/genetics , Insulin Secretion , Intracellular Signaling Peptides and Proteins , Mice , Mice, Knockout , Proteins/genetics , trans-Golgi Network/metabolism
14.
J Neurochem ; 129(2): 328-38, 2014 Apr.
Article En | MEDLINE | ID: mdl-24345054

Gain-of-toxic-function mutations in Seipin (Asparagine 88 to Serine (N88S) and Serine 90 to Leucine (S90L) mutations, both of which disrupt the N-glycosylation) cause autosomal dominant motor neuron diseases. However, the mechanism of how these missense mutations lead to motor neuropathy is unclear. Here, we analyze the impact of disruption of N-glycosylation of Seipin on synaptic transmission by over-expressing mutant Seipin in cultured cortical neurons via lentiviral infection. Immunostaining shows that over-expressed Seipin is partly colocalized with synaptic vesicle marker synaptophysin. Electrophysiological recordings reveal that the Seipin mutation significantly decreases the frequency, but not the amplitudes of miniature excitatory post-synaptic currents and miniature inhibitory post-synaptic currents. The amplitude of both evoked excitatory post-synaptic currents and inhibitory post-synaptic current is also compromised by mutant Seipin over-expression. The readily releasable pool and vesicular release probability of synaptic vesicles are both altered in neurons over-expressing Seipin-N88S, whereas neither γ-amino butyric acid (GABA) nor α-Amino-3-hydroxy-5-methyl-4- isoxazolepropionic acid (AMPA) induced whole cell currents are affected. Moreover, electron microscopy analysis reveals decreased number of morphologically docked synaptic vesicles in Seipin-N88S-expressing neurons. These data demonstrate that Seipin-N88S mutation impairs synaptic neurotransmission, possibly by regulating the priming and docking of synaptic vesicles at the synapse.


Heterotrimeric GTP-Binding Proteins/genetics , Heterotrimeric GTP-Binding Proteins/physiology , Motor Neuron Disease/genetics , Mutation, Missense/genetics , Mutation, Missense/physiology , Synaptic Transmission/genetics , Synaptic Transmission/physiology , Animals , Cells, Cultured , Excitatory Postsynaptic Potentials/physiology , Female , GTP-Binding Protein gamma Subunits , Gene Expression/drug effects , Gene Expression/physiology , Genetic Vectors , Image Processing, Computer-Assisted , Immunohistochemistry , Lentivirus/genetics , Male , Mice , Mice, Inbred C57BL , Microscopy, Electron, Transmission , Spastic Paraplegia, Hereditary/genetics , Spastic Paraplegia, Hereditary/physiopathology , Synapses/physiology
15.
J Neurochem ; 124(4): 478-89, 2013 Feb.
Article En | MEDLINE | ID: mdl-23173741

Heterozygosity for missense mutations in Seipin, namely N88S and S90L, leads to a broad spectrum of motor neuropathy, while a number of loss-of-function mutations in Seipin are associated with the Berardinelli-Seip congenital generalized lipodystrophy type 2 (CGL2, BSCL2), a condition that is characterized by severe lipoatrophy, insulin resistance, and intellectual impairment. The mechanisms by which Seipin mutations lead to motor neuropathy, lipodystrophy, and insulin resistance, and the role Seipin plays in central nervous system (CNS) remain unknown. The goal of this study is to understand the functions of Seipin in the CNS using a loss-of-function approach, i.e. by knockdown (KD) of Seipin gene expression. Excitatory post-synaptic currents (EPSCs) were impaired in Seipin-KD neurons, while the inhibitory post-synaptic currents (IPSCs) remained unaffected. Expression of a shRNA-resistant human Seipin rescued the impairment of EPSC produced by Seipin KD. Furthermore, α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA)-induced whole-cell currents were significantly reduced in Seipin KD neurons, which could be rescued by expression of a shRNA-resistant human Seipin. Fluorescent imaging and biochemical studies revealed reduced level of surface AMPA receptors, while no obvious ultrastructural changes in the pre-synapse were found. These data suggest that Seipin regulates excitatory synaptic function through a post-synaptic mechanism.


Cerebral Cortex/cytology , Heterotrimeric GTP-Binding Proteins/metabolism , Mutation, Missense/genetics , Neurons/physiology , Synaptic Transmission/genetics , 6-Cyano-7-nitroquinoxaline-2,3-dione/pharmacology , Animals , Biotinylation , Brain/cytology , Brain/metabolism , Cells, Cultured , Dose-Response Relationship, Drug , Enzyme-Linked Immunosorbent Assay , Excitatory Amino Acid Agents/pharmacology , Excitatory Postsynaptic Potentials/drug effects , Excitatory Postsynaptic Potentials/genetics , GTP-Binding Protein gamma Subunits , Gene Expression Regulation/drug effects , Gene Expression Regulation/genetics , Heterotrimeric GTP-Binding Proteins/genetics , Immunoprecipitation , Lipodystrophy, Congenital Generalized , Mice , Microscopy, Electron, Transmission , Neurons/drug effects , Neurons/ultrastructure , Patch-Clamp Techniques , RNA, Small Interfering/metabolism , RNA, Small Interfering/pharmacology , Receptors, AMPA/metabolism , Synapses/metabolism , Synapses/ultrastructure , Synaptic Transmission/drug effects , Synaptophysin/metabolism , Synaptosomes/drug effects , Synaptosomes/metabolism , Synaptosomes/ultrastructure , alpha-Amino-3-hydroxy-5-methyl-4-isoxazolepropionic Acid/pharmacology
16.
Tissue Eng Part C Methods ; 19(2): 166-80, 2013 Feb.
Article En | MEDLINE | ID: mdl-22834957

Neural progenitor cells (NPCs) derived from human induced pluripotent stem cells (hiPSCs) can be differentiated to neural cells that model neurodegenerative diseases and be used in the screening of potential drugs to ameliorate the disease phenotype. Traditionally, NPCs are produced in 2D cultures, in low yields, using a laborious process that includes generation of embryonic bodies, plating, and colony selections. To simplify the process and generate large numbers of hiPSC-derived NPCs, we introduce a microcarrier (MC) system for the expansion of a hiPSC line and its subsequent differentiation to NPC, using iPS (IMR90) as a model cell line. In the expansion stage, a process of cell propagation in serum-free MC culture was developed first in static culture, which is then scaled up in stirred spinner flasks. A 7.7-fold expansion of iPS (IMR90) and cell yield of 1.3×106 cells/mL in 7 days of static MC culture were achieved. These cells maintained expression of OCT 3/4 and TRA-1-60 and possessed a normal karyotype over 10 passages. A higher cell yield of 6.1×106 cells/mL and 20-fold hiPSC expansion were attained using stirred spinner flasks (seeded from MC static cultures) and changing the medium-exchange regimen from once to twice a day. In the differentiation stage, NPCs were generated with 78%-85% efficiency from hiPSCs using a simple serum-free differentiation protocol. Finally, the integrated process of cell expansion and differentiation of hiPSCs into NPCs using an MC in spinner flasks yielded 333 NPCs per seeded hiPSC as compared to 53 in the classical 2D tissue culture protocol. Similar results were obtained with the HES-3 human embryonic stem cell line. These NPCs were further differentiated into ßIII-tubulin⁺ neurons, GFAP⁺ astrocytes, and O4⁺ oligodendrocytes, showing that cells maintained their multilineage differentiation potential.


Cell Differentiation , Cell Division , Neurons/cytology , Pluripotent Stem Cells/cytology , Cells, Cultured , Culture Media, Serum-Free , Flow Cytometry , Humans , Immunohistochemistry , Karyotyping , Real-Time Polymerase Chain Reaction
17.
Stem Cells Dev ; 21(5): 729-41, 2012 Mar 20.
Article En | MEDLINE | ID: mdl-21649559

Molecular and cellular signaling pathways are involved in the process of neural differentiation from human embryonic stem cells (hESC) to terminally differentiated neurons. The Sonic hedgehog (SHH) morphogen is required to direct the differentiation of hESC to several neural subtypes, for example, dopaminergic (DA) or motor neurons. However, the roles of SHH signaling and the pathway target genes that regulate the diversity of cellular responses arising from SHH activation during neurogenesis of hESC have yet to be elucidated. In this study, we report that overexpression of SHH in hESC promotes the derivation of neuroprogenitors (NP), increases proliferation of NP, and subsequently increases the yield of DA neurons. Next, gene expression changes resulting from the overexpression of SHH in hESC-derived NP were examined by genome-wide transcriptional profiling. Categorizing the differentially expressed genes according to the Gene Ontology biological processes showed that they are involved in numerous cellular processes, including neural development, NP proliferation, and neural specification. In silico GLI-binding sites analysis of the differentially expressed genes also identified a set of putative novel direct target genes of SHH in hESC-derived NP, which are involved in nervous system development. Electrophoretic mobility shift assays and promoter-luciferase assays confirmed that GLI1 binds to the promoter region and activates transcription of HEY2, a NOTCH signaling target gene. Taken together, our data provide evidence for the first time that there is cross-talk between the NOTCH and SHH signaling pathways in hESC-derived NP and also provide significant new insights into transcriptional targets in SHH-mediated neural differentiation of hESC.


Dopaminergic Neurons/metabolism , Embryonic Stem Cells/metabolism , Gene Expression Profiling , Hedgehog Proteins/genetics , Neural Stem Cells/metabolism , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Blotting, Western , Cell Differentiation/genetics , Cell Line , Dopaminergic Neurons/cytology , Dopaminergic Neurons/physiology , Embryonic Stem Cells/cytology , Eye Proteins/genetics , Eye Proteins/metabolism , Fluorescent Antibody Technique , Glial Fibrillary Acidic Protein/genetics , Glial Fibrillary Acidic Protein/metabolism , Hedgehog Proteins/metabolism , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , Intermediate Filament Proteins/genetics , Intermediate Filament Proteins/metabolism , Membrane Potentials , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Nestin , Neural Stem Cells/cytology , Neural Stem Cells/physiology , PAX6 Transcription Factor , Paired Box Transcription Factors/genetics , Paired Box Transcription Factors/metabolism , Patch-Clamp Techniques , Promoter Regions, Genetic/genetics , Protein Binding , Repressor Proteins/genetics , Repressor Proteins/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Transcription Factors/genetics , Transcription Factors/metabolism , Zinc Finger Protein GLI1
18.
Mol Cell Neurosci ; 45(4): 418-29, 2010 Dec.
Article En | MEDLINE | ID: mdl-20696250

EHD1 is an EH (Eps15 homology) domain-containing protein involved in endosomal recycling. Our yeast two hybrid screening experiments showed that EHD1 interacts with a synaptic protein, snapin, and the present study was carried out to further elucidate the functional significance of this interaction. Immunoreactivity to EHD1 is observed in the cerebral cortex, hippocampus and striatum, in the rat brain. The protein is colocalized with the axon terminal marker synaptophysin in cultured neurons. EHD1 binds to the C terminus of snapin via its C terminus EH domain. It negatively affects the binding of a SNARE complex protein, SNAP-25, to snapin, probably due to the competition for overlapping binding sites on the C terminus of snapin. EHD1 affects the coupling of synaptotagmin-1 to the SNARE complex, and could be a negative regulator of exocytosis. This is supported by electrophysiological findings that PC-12 cells which overexpress EHD1 show reduced depolarization-induced exocytosis compared to controls, but the reduced exocytosis is not observed in cells which overexpress the N terminus of EHD1 that is unable to bind snapin. Together, the above results indicate that EHD1 is a synaptic protein that negatively affects exocytosis through binding to snapin.


Exocytosis/physiology , Neurons/metabolism , Synapses/metabolism , Vesicular Transport Proteins/metabolism , Animals , Brain/metabolism , Immunoprecipitation , Microscopy, Immunoelectron , PC12 Cells , Rats , SNARE Proteins/metabolism , Transfection , Two-Hybrid System Techniques
19.
J Physiol ; 587(Pt 6): 1169-78, 2009 Mar 15.
Article En | MEDLINE | ID: mdl-19171650

Hormones such as glucagon are secreted by Ca(2+)-induced exocytosis of large dense-core vesicles, but the mechanisms involved have only been partially elucidated. Studies of pancreatic beta-cells secreting insulin revealed that synaptotagmin-7 alone is not sufficient to mediate Ca(2+)-dependent insulin granule exocytosis, and studies of chromaffin cells secreting neuropeptides and catecholamines showed that synaptotagmin-1 and -7 collaborate as Ca(2+) sensors for exocytosis, and that both are equally involved. As no other peptide secretion was analysed, it remains unclear whether synaptotagmins generally act as Ca(2+) sensors in large dense-core vesicle exocytosis in endocrine cells, and if so, whether synaptotagmin-7 always functions with a partner in that role. In particular, far less is known about the mechanisms underlying Ca(2+)-triggered glucagon release from alpha-cells than insulin secretion from beta-cells, even though insulin and glucagon together regulate blood glucose levels. To address these issues, we analysed the role of synaptotagmins in Ca(2+)-triggered glucagon exocytosis. Surprisingly, we find that deletion of a single synaptotagmin isoform, synaptotagmin-7, nearly abolished Ca(2+)-triggered glucagon secretion. Moreover, single-cell capacitance measurements confirmed that pancreatic alpha-cells lacking synaptotagmin-7 exhibited little Ca(2+)-induced exocytosis, whereas all other physiological and morphological parameters of the alpha-cells were normal. Our data thus identify synaptotagmin-7 as a principal Ca(2+) sensor for glucagon secretion, and support the notion that synaptotagmins perform a universal but selective function as individually acting Ca(2+) sensors in neurotransmitter, neuropeptide, and hormone secretion.


Exocytosis/physiology , Glucagon-Secreting Cells/metabolism , Glucagon/metabolism , Intracellular Calcium-Sensing Proteins/physiology , Synaptotagmins/physiology , Action Potentials/physiology , Animals , Blood Glucose/drug effects , Calcium Channels/metabolism , Exocytosis/drug effects , Gene Expression/genetics , Glucagon/blood , Glucagon/genetics , Glucagon/pharmacology , Glucagon-Secreting Cells/ultrastructure , Hypoglycemia/blood , Insulin/pharmacology , Islets of Langerhans/metabolism , Male , Mice , Mice, Inbred Strains , Mice, Knockout , omega-Conotoxins/pharmacology
20.
Endocrinology ; 150(5): 2127-35, 2009 May.
Article En | MEDLINE | ID: mdl-19116346

Insulin secretion from beta-cells is regulated by a complex signaling network. Our earlier study has reported that Rac1 participates in glucose- and cAMP-induced insulin secretion probably via maintaining a functional actin structure for recruitment of insulin granules. Type Ialpha phosphatidylinositol-4-phosphate 5-kinase (PIP5K-Ialpha) is a downstream effector of Rac1 and a critical enzyme for synthesis of phosphatidylinositol-4,5-bisphosphate (PIP(2)). By using an RNA interference technique, PIP5K-Ialpha in INS-1 beta-cells could be specifically knocked down by 70-75%. PIP5K-Ialpha knockdown disrupted filamentous actin structure and caused changes in cell morphology. In addition, PIP(2) content in the plasma membrane was reduced and the glucose effect on PIP(2) was abolished but without affecting glucose-induced formation of inositol 1,4,5-trisphosphate. At basal conditions (2.8 mM glucose), PIP5K-Ialpha knockdown doubled insulin secretion, elevated glucose metabolic rate, depolarized resting membrane potential, and raised cytoplasmic free Ca(2+) levels ([Ca(2+)](i)). The total insulin release at high glucose was increased upon PIP5K-Ialpha knockdown. However, the percent increment of insulin secretion by high glucose and forskolin over the basal release was significantly reduced, an effect more apparent on the late phase of insulin secretion. Metabolism and [Ca(2+)](i) rises at high glucose were also attenuated in cells after PIP5K-Ialpha knockdown. In contrast, PIP5K-Ialpha knockdown had no effect on cell growth and viability. Taken together, our data suggest that PIP5K-Ialpha may play an important role in both the proximal and distal steps of signaling cascade for insulin secretion in beta-cells.


Glucose/metabolism , Insulin-Secreting Cells/physiology , Insulin/metabolism , Membrane Potentials/genetics , Phosphotransferases (Alcohol Group Acceptor)/physiology , Actins/metabolism , Animals , Cell Death/drug effects , Cell Death/genetics , Cell Line , Cell Proliferation/drug effects , Cell Shape , Gene Knockdown Techniques , Glucose/pharmacology , Insulin Secretion , Insulin-Secreting Cells/cytology , Insulin-Secreting Cells/drug effects , Insulin-Secreting Cells/metabolism , Mice , Phosphatidylinositol 4,5-Diphosphate/metabolism , Phosphotransferases (Alcohol Group Acceptor)/antagonists & inhibitors , Phosphotransferases (Alcohol Group Acceptor)/genetics , RNA, Small Interfering/genetics , RNA, Small Interfering/pharmacology , Signal Transduction/drug effects , Signal Transduction/genetics , Transfection
...