Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 43
Filter
1.
Cell Biol Int ; 44(1): 343-351, 2020 Jan.
Article in English | MEDLINE | ID: mdl-31498530

ABSTRACT

Neuroinflammatory disorders such as Alzheimer's and Parkinson's diseases are characterised by chronic inflammation and loss of vascular integrity. Bradykinin 1 receptor (B1R) activation has been implicated in many neuroinflammatory diseases, but the contribution of B1R to inflammation and vascular breakdown is yet to be determined. As a result, the present study evaluated the effect of B1R stimulation using Des-Arg-9-BK on the cytokine profile and junctional properties of human cerebral microvascular endothelial cells (hCMVECs). Results showed that stimulation of B1R receptors increased secretion of pro-inflammatory cytokines, interleukin-6 (IL-6), IL-8, intracellular adhesion molecule-1 (ICAM-1), vascular cell adhesion molecule-1 (VCAM-1) and monocyte chemoattractant protein-1 (MCP-1), but decreased the expression of vascular endothelial growth factor (VEGF), a cytokine and growth factor required for maintenance of the vasculature. B1R stimulation also resulted in the loss of occludin expression at tight junctions with no change in VE-cadherin expression. There was also a significant increase in permeability to Evans blue albumin, suggesting an increase of vascular permeability. Taken together, these results suggest that B1R activation that occurs in neuroinflammatory diseases may contribute to both the inflammation and loss of blood-brain barrier integrity that is characteristic of these diseases.

2.
Biochim Biophys Acta Biomembr ; 1860(1): 224-236, 2018 Jan.
Article in English | MEDLINE | ID: mdl-28347700

ABSTRACT

A common cause of mortality and long-term adult disability, cerebral ischemia or brain ischemia imposes a significant health and financial burden on communities worldwide. Cerebral ischemia is a condition that arises from a sudden loss of blood flow and consequent failure to meet the high metabolic demands of the brain. The lack of blood flow initiates a sequelae of cell death mechanisms, including the activation of the inflammatory pathway, which can ultimately result in irreversible brain tissue damage. In particular, Connexins and Pannexins are non-selective channels with a large pore that have shown to play time-dependent roles in the perpetuation of ischaemic injury. This review highlights the roles of Connexin and Pannexin channels in cell death mechanisms as a promising therapeutic target in cerebral ischemia, and in particular connexin hemichannels which may contribute most of the ATP release as a result of ischemia as well as during reperfusion. This article is part of a Special Issue entitled: Gap Junction Proteins edited by Jean Claude Herve.


Subject(s)
Adenosine Triphosphate/metabolism , Brain Ischemia/metabolism , Cerebral Cortex/metabolism , Connexins/metabolism , Ion Channels/metabolism , Animals , Brain Ischemia/pathology , Cerebral Cortex/pathology , Humans
3.
Biochim Biophys Acta Gen Subj ; 1861(2): 68-78, 2017 Feb.
Article in English | MEDLINE | ID: mdl-27816754

ABSTRACT

BACKGROUND: Non-selective Connexin43 hemichannels contribute to secondary lesion spread. The hemichannel blocking peptidomimetic Peptide5, derived from the second extracellular loop of the human Connexin43 protein, prevents lesion spread and reduces vascular permeability in preclinical models of central nervous system injury. The molecular mode of action of Peptide5, however, was unknown and is described here. METHODS: Human cerebral microvascular endothelial cells and APRE-19 cells were used. Scrape loading was used to assess gap junction function and hypoxic, acidic ion-shifted Ringer solution induced ATP release used to assess hemichannel function. Peptide modifications, including amino acid substitutions and truncations, and competition assays were used to demonstrate Peptide5 functional specificity and site of action respectively. RESULTS: Peptide5 inhibits Connexin43 hemichannel-mediated ATP release by acting on extracellular loop two of Connexin43, adjacent to its matching sequence within the protein. Precise sequence specificity is important for hemichannel block, but less so for uncoupling of gap junction channels (seen only at high concentrations). The SRPTEKT motif is central to Peptide5 function but on its own is not sufficient to inhibit hemichannels. Both the SRPTEKT motif and Peptide5 reduce gap junction communication, but neither uncoupling below 50%. CONCLUSIONS: Reduced gap junction coupling at high peptide concentrations appears to be relatively non-specific. However, Peptide5 at low concentrations acts upon extracellular loop two of Connexin43 to block hemichannels in a precise, sequence specific manner. GENERAL SIGNIFICANCE: The concentration dependent and sequence specific action of Peptide5 supports its development for the treatment of retinal injury and chronic disease, as well as other central nervous system injury and disease conditions.


Subject(s)
Cerebrum/drug effects , Connexin 43/metabolism , Endothelial Cells/drug effects , Ischemia/drug therapy , Peptides/pharmacology , Reperfusion Injury/drug therapy , Adenosine Triphosphate/metabolism , Cell Line , Central Nervous System Diseases/drug therapy , Central Nervous System Diseases/metabolism , Cerebrum/metabolism , Endothelial Cells/metabolism , Gap Junctions/drug effects , Gap Junctions/metabolism , Humans , Ion Channels/metabolism , Ischemia/metabolism , Reperfusion Injury/metabolism , Retinal Diseases/drug therapy , Retinal Diseases/metabolism
4.
Exp Brain Res ; 235(10): 3033-3048, 2017 10.
Article in English | MEDLINE | ID: mdl-28725925

ABSTRACT

Systemic administration of a Connexin43 mimetic peptide, Peptide5, has been shown to reduce secondary tissue damage and improve functional recovery after spinal cord injury (SCI). This study investigated safety measures and potential off-target effects of Peptide5 systemic administration. Rats were subjected to a mild contusion SCI using the New York University impactor. One cohort was injected intraperitoneally with a single dose of fluorescently labelled Peptide5 and euthanised at 2 or 4 h post-injury for peptide distribution analysis. A second cohort received intraperitoneal injections of Peptide5 or a scrambled peptide and was culled at 8 or 24 h post-injury for the analysis of connexin proteins and systemic cytokine profile. We found that Peptide5 did not cross the blood-spinal cord barrier in control animals, but reached the lesion area in the spinal cord-injured animals without entering non-injured tissue. There was no evidence that the systemic administration of Peptide5 modulates Connexin43 protein expression or hemichannel closure in the heart and lung tissue of SCI animals. The expression levels of other major connexin proteins including Connexin30 in astrocytes, Connexin36 in neurons and Connexin47 in oligodendrocytes were also unaltered by systemic delivery of Peptide5 in either the injured or non-injured spinal cords. In addition, systemic delivery of Peptide5 had no significant effect on the plasma levels of cytokines, chemokines or growth factors. These data indicate that the systemic delivery of Peptide5 is unlikely to cause any off-target or adverse effects and may thus be a safe treatment option for traumatic SCI.


Subject(s)
Biomimetic Materials/pharmacology , Connexin 43/pharmacology , Spinal Cord Injuries/drug therapy , Animals , Biomimetic Materials/administration & dosage , Biomimetic Materials/adverse effects , Biomimetic Materials/pharmacokinetics , Connexin 43/administration & dosage , Connexin 43/adverse effects , Connexin 43/pharmacokinetics , Disease Models, Animal , Female , Rats , Rats, Sprague-Dawley
5.
J Neurochem ; 134(5): 927-42, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26011179

ABSTRACT

Huntington's disease (HD) is an autosomal dominant neurodegenerative disease caused by an expansion of the CAG repeat in the huntingtin gene. One of the brain changes that occurs in HD is the expression of the receptor for advanced glycation end products (RAGE), a receptor protein capable of activating multiple signalling pathways by interacting with a range of ligands leading to either beneficial or harmful effects to the cell. Here, we demonstrate in human HD brains a high degree of co-localization of RAGE with its putative ligands S100B and N-carboxymethyllysine (CML) in the caudate nucleus (CN) and the subependymal layer (SEL). The level of co-staining for both RAGE-S100B and RAGE-CML was the highest in the astrocytes but was low in neurons and microglia. The immunostaining for RAGE, S100B and CML extended in a medio-lateral (SEL-CN) direction with increasing grade, such that any change in the expression and co-localization pattern between grades was less prominent in the lateral CN. Additionally, signalling molecules that are downstream of RAGE activation showed changes in their activation status in HD brains. A larger number of RAGE-positive astrocytic cells had NF-kB translocated to the nucleus and the level of phospho-ERK1/2 was also increased in HD brains. Interestingly, the level of mDia-1, that interacts directly with the cytoplasmic domain of RAGE, decreased in HD. Overall, the results suggest a correlation between the functions of RAGE and the HD pathology, but the influence of RAGE on astrocytes and the impact of this on HD progression requires further study. RAGE (receptor for advanced glycation end products) binds multiple types of ligand to produce either neurotrophic or neurotoxic effects. Immunohistochemical staining of HD human brains showed that both RAGE and its ligands were expressed primarily in astrocytes. The pattern of staining corresponded to the grade and region-wise pattern of neurodegeneration suggesting a possible role for RAGE in HD pathology.


Subject(s)
Astrocytes/metabolism , Corpus Striatum/metabolism , Huntington Disease/metabolism , Lysine/analogs & derivatives , Nerve Tissue Proteins/metabolism , Receptors, Immunologic/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Aged , Aged, 80 and over , Corpus Striatum/pathology , Ependyma , Female , Formins , Humans , Huntington Disease/pathology , Lysine/metabolism , MAP Kinase Signaling System , Male , Microglia/metabolism , Middle Aged , NF-kappa B/metabolism , Neurons/metabolism , Receptor for Advanced Glycation End Products , S100 Calcium Binding Protein beta Subunit/metabolism , Severity of Illness Index
6.
Ann Neurol ; 71(1): 121-32, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22275258

ABSTRACT

OBJECTIVE: Connexin hemichannels can open during ischemia, resulting in loss of membrane potential, calcium influx, and release of glutamate. In this study, we tested the hypothesis that opening of hemichannels after cerebral ischemia may contribute to delayed evolution of injury. METHODS: We infused a mimetic peptide that blocks connexin 43 hemichannels into the lateral ventricle of chronically instrumented fetal sheep in utero at 128 ± 1 days gestation (term is 147 days), starting 90 minutes after 30 minutes of severe ischemia induced by reversible bilateral carotid artery occlusion, for either 1 or 25 hours. Sheep were killed 7 days later. RESULTS: Peptide infusion was associated with a graded improvement in recovery of electroencephalographic power after 7 days recovery, from -13 ± 1.9 dB (n = 7) after ischemia-vehicle to -9 ± 1.6 dB (n = 7) after ischemia-short infusion and -5 ± 1.6 dB after ischemia-long infusion (n = 6, p < 0.05). Peptide infusion was associated with reduced seizure activity after ischemia, less frequent status epilepticus (p < 0.05), and earlier return of sleep state cycling (p < 0.05). Ischemia-long infusion (but not ischemia-short infusion) was associated with improved survival of oligodendrocytes in intragyral and periventricular white matter (p < 0.05) and increased brain weight (p < 0.05). Ischemia-long infusion was associated with an intermediate estimate of surviving neurons in the parasagittal cortex of 2.9 ± 0.8 × 10(6), in comparison to sham control (4.3 ± 0.9 × 10(6)) or ischemia-vehicle (1.5 ± 0.4 × 10(6); p < 0.05 vs sham control). INTERPRETATION: These data support the hypothesis that opening of connexin hemichannels is a significant mediator of postischemic white and gray matter dysfunction and injury.


Subject(s)
Brain Ischemia/metabolism , Brain Ischemia/prevention & control , Connexin 43/antagonists & inhibitors , Fetal Hypoxia/drug therapy , Peptide Fragments/administration & dosage , Animals , Brain Ischemia/physiopathology , Cell Survival/physiology , Connexin 43/physiology , Disease Models, Animal , Electroencephalography , Female , Fetal Hypoxia/metabolism , Fetal Hypoxia/pathology , Gene Targeting , Male , Neurons/pathology , Neurons/physiology , Peptide Fragments/chemical synthesis , Peptide Fragments/therapeutic use , Pregnancy , Seizures/prevention & control , Sheep , Sleep Stages/physiology , Treatment Outcome
7.
Brain ; 135(Pt 2): 506-20, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22345088

ABSTRACT

Connexin43 gap junction protein is expressed in astrocytes and the vascular endothelium in the central nervous system. It is upregulated following central nervous system injury and is recognized as playing an important role in modulating the extent of damage. Studies that have transiently blocked connexin43 in spinal cord injury and central nervous system epileptic models have reported neuronal rescue. The purpose of this study was to investigate neuronal rescue following retinal ischaemia-reperfusion by transiently blocking connexin43 activity using a connexin43 mimetic peptide. A further aim was to evaluate the effect of transiently blocking connexin43 on vascular permeability as this is known to increase following central nervous system ischaemia. Adult male Wistar rats were exposed to 60 min of retinal ischaemia. Treatment groups consisted of no treatment, connexin43 mimetic peptide and scrambled peptide. Retinas were then evaluated at 1-2, 4, 8 and 24 h, and 7 and 21 days post-ischaemia. Evans blue dye leak from retinal blood vessels was used to assess vascular leakage. Blood vessel integrity was examined using isolectin-B4 labelling. Connexin43 levels and astrocyte activation (glial fibrillary acidic protein) were assessed using immunohistochemistry and western blot analysis. Retinal whole mounts and retinal ganglion cell counts were used to quantify neurodegeneration. An in vitro cell culture model of endothelial cell ischaemia was used to assess the effect of connexin43 mimetic peptide on endothelial cell survival and connexin43 hemichannel opening using propidium iodide dye uptake. We found that retinal ischaemia-reperfusion induced significant vascular leakage and disruption at 1-2, 4 and 24 h following injury with a peak at 4 h. Connexin43 immunoreactivity was significantly increased at 1-2, 4, 8 and 24 h post ischaemia-reperfusion injury co-localizing with activated astrocytes, Muller cells and vascular endothelial cells. Connexin43 mimetic peptide significantly reduced dye leak at 4 and 24 h. In vitro studies on endothelial cells demonstrate that endothelial cell death following hypoxia can be mediated directly by opening of connexin43 hemichannels in endothelial cells. Blocking connexin43 mediated vascular leakage using a connexin43 mimetic peptide led to increased retinal ganglion cell survival at 7 and 21 days to levels of uninjured retinas. Treatment with scrambled peptide did not result in retinal ganglion cell rescue. Pharmacological targeting of connexin43 gap junction protein by transiently blocking gap junction hemichannels following injury provides new opportunities for treatment of central nervous system ischaemia.


Subject(s)
Connexin 43/antagonists & inhibitors , Ischemia/drug therapy , Oligopeptides/therapeutic use , Retina/drug effects , Retinal Diseases/drug therapy , Retinal Ganglion Cells/drug effects , Retinal Vessels/drug effects , Animals , Connexin 43/metabolism , Ischemia/metabolism , Ischemia/pathology , Male , Rats , Rats, Wistar , Retina/metabolism , Retina/pathology , Retinal Diseases/metabolism , Retinal Diseases/pathology , Retinal Ganglion Cells/metabolism , Retinal Ganglion Cells/pathology , Retinal Vessels/metabolism , Retinal Vessels/pathology
8.
J Neurosci Res ; 90(6): 1136-47, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22344976

ABSTRACT

The receptor for advanced glycation end products (RAGE) is a multiligand receptor protein thought to play an important role in neuronal differentiation. RAGE can bind a number of ligands and activate a variety of signalling pathways that lead to diverse downstream effects. Amphoterin and S100B are endogenous ligands, the interaction of which with RAGE is known to be involved in defined physiological processes. The present study investigated the spatiotemporal pattern of the expression for RAGE and its ligands, amphoterin and S100B, during neuronal differentiation of NT2/D1 cells. In this study, all three proteins were shown to increase with progression of neuronal differentiation as determined by Western blotting, raising the possibility that both amphoterin and S100B may interact with RAGE and have important functions during the process of cell differentiation. Moreover, blocking the activation of RAGE with neutralizing antibody in the presence of retinoic acid disrupted the progression of normal neuronal differentiation. Immunocytochemistry (ICC) studies showed that amphoterin partially colocalized with RAGE within differentiating NT2 cells, whereas S100B showed a high degree of colocalization. This result suggests that S100B is more likely to be the principal ligand for RAGE during the differentiation process and that RAGE and amphoterin might have both independent and combined roles. Moreover, RAGE was expressed only in cells that were committed to a neuronal phenotype, suggesting direct involvement of RAGE in mediating cellular changes within differentiating neuronal cells. Further detailed studies are now required to characterize fully the role of RAGE during the neuronal differentiation period.


Subject(s)
Cell Differentiation/physiology , Neurons/physiology , Receptors, Immunologic/physiology , Antibodies/pharmacology , Antineoplastic Agents/pharmacology , Carcinoma/pathology , Cell Cycle/drug effects , Cell Differentiation/drug effects , Cell Line, Tumor , HMGB1 Protein/genetics , HMGB1 Protein/metabolism , Humans , Nerve Growth Factors/genetics , Nerve Growth Factors/metabolism , Neurons/drug effects , Receptor for Advanced Glycation End Products , Receptors, Immunologic/immunology , S100 Calcium Binding Protein beta Subunit , S100 Proteins/genetics , S100 Proteins/metabolism , Tretinoin/pharmacology , Up-Regulation/drug effects , Up-Regulation/physiology
9.
Anal Biochem ; 431(2): 90-5, 2012 Dec 15.
Article in English | MEDLINE | ID: mdl-22929700

ABSTRACT

Gene expression studies often require inducible coexpression of both a gene of interest and a reporter gene. Fusion of fluorescent reporters can, however, modify protein structure and function. We have generated inducible expression systems for two connexin genes: Cx30 and Cx43. It has been reported recently that reporter fusion to connexins can modify their function. Therefore, we compared two methods of independent reporter coexpression and examined colocalization with induced connexin expression. Identical levels of connexin expression were observed for both the bidirectional and bicistronic expression systems. In contrast, however, reporter gene expression by the bidirectional promoter provided brighter average fluorescent pixel intensity than expression of a reporter gene in a bicistronic transcript. Moreover, as a result of this difference in reporter expression, bidirectional expression systems provided equal or better colocalization between the connexins and reporter gene fluorescence. The results of our study indicate that bidirectional reporter expression provides a robust indicator of transfection and gene expression and, therefore, may favor the use of bidirectional over bicistronic reporters in the design of expression systems where the gene of interest, such as a connexin gene, contains translational motifs or long intronic regions.


Subject(s)
Connexin 43 , Connexins , Gene Expression , Genes, Reporter , Connexin 30 , Connexin 43/genetics , Connexin 43/metabolism , Connexins/genetics , Connexins/metabolism , Green Fluorescent Proteins/genetics , Humans , Promoter Regions, Genetic
10.
Int J Mol Sci ; 13(5): 6303-6319, 2012.
Article in English | MEDLINE | ID: mdl-22754366

ABSTRACT

Hypoxic-ischaemic brain injury at birth is associated with 1-3/1000 cases of moderate to severe encephalopathy. Previously, we have shown that connexin 43 hemichannel blockade, with a specific mimetic peptide, reduced the occurrence of seizures, improved recovery of EEG power and sleep state cycling, and improved cell survival following global cerebral ischaemia. In the present study, we examined the dose response for intracerebroventricular mimetic peptide infusion (50 µmol/kg/h for 1 h, followed by 50 µmol/kg/24 h (low dose) or 50 µmol/kg/h for 25 h (high dose) or vehicle only (control group), starting 90 min after the end of ischaemia), following global cerebral ischaemia, induced by 30 min bilateral carotid artery occlusion, in near-term fetal sheep (128 ± 1 days gestation). Both peptide infusion groups were associated with a transient significant increase in EEG power between 2-12 h after ischaemia. The ischaemia-low dose group showed a significant recovery of EEG power from day five compared to the ischaemia-vehicle and -high dose groups. In contrast, the high dose infusion was associated with greater secondary increase in impedance (brain cell swelling), as well as a trend towards a greater increase in lactate concentration and mortality. These data suggest that higher doses of connexin mimetic peptide are not beneficial and may be associated with adverse outcomes, most likely attributable to uncoupling of connexin 43 gap junctions leading to dysfunction of the astrocytic syncytium.


Subject(s)
Brain/physiopathology , Connexin 43/metabolism , Fetal Hypoxia/veterinary , Hypoxia-Ischemia, Brain/veterinary , Oligopeptides/adverse effects , Sheep Diseases/drug therapy , Animals , Biomimetics , Brain/drug effects , Brain/embryology , Disease Models, Animal , Dose-Response Relationship, Drug , Electroencephalography/drug effects , Female , Fetal Hypoxia/drug therapy , Fetal Hypoxia/physiopathology , Hypoxia-Ischemia, Brain/drug therapy , Hypoxia-Ischemia, Brain/physiopathology , Infusions, Intraventricular , Oligopeptides/administration & dosage , Pregnancy , Seizures/drug therapy , Seizures/physiopathology , Seizures/veterinary , Sheep , Sheep Diseases/physiopathology
11.
Int J Neurosci ; 120(12): 752-9, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20942591

ABSTRACT

A reliable model system of epileptiform insult would facilitate investigation into the underlying biological mechanisms. Epileptiform insult was induced in hippocampal slice cultures by lowering extracellular Mg(2+), (+)-bicuculline, or (-)-bicuculline methochloride, a stable salt form of bicuculline (both forms block GABA(A) receptors). Cell death was assessed by propidium iodide uptake. Low Mg(2+) or (+)-bicuculline did not produce cell death regardless of dose or incubation period. Exposure to 100 microM (-)-bicuculline methochloride for 48 hr resulted in prominent CA1 cell death. These findings demonstrate that not all pro-epileptic drugs/ion changes used routinely for electrophysiological recording of seizure activity lead to cell death in hippocampal slice cultures and that treatment with bicuculline methochloride can be used as a reliable model for epileptiform insult.


Subject(s)
Bicuculline/toxicity , Hippocampus/metabolism , Hippocampus/pathology , Magnesium/metabolism , Animals , Animals, Newborn , Cell Death/drug effects , Cell Death/physiology , Convulsants/toxicity , Hippocampus/drug effects , Organ Culture Techniques , Rats , Rats, Wistar
12.
Eur J Neurosci ; 30(10): 1849-59, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19912331

ABSTRACT

Rotenone is a toxin used to generate animal models of Parkinson's disease; however, the mechanisms of toxicity in substantia nigra pars compacta (SNc) neurons have not been well characterized. We have investigated rotenone (0.05-1 microm) effects on SNc neurons in acute rat midbrain slices, using whole-cell patch-clamp recording combined with microfluorometry. Rotenone evoked a tolbutamide-sensitive outward current (94 +/- 15 pA) associated with increases in intracellular [Ca(2+)] ([Ca(2+)](i)) (73.8 +/- 7.7 nm) and intracellular [Na(+)] (3.1 +/- 0.6 mm) (all with 1 microm). The outward current was not affected by a high ATP level (10 mm) in the patch pipette but was decreased by Trolox. The [Ca(2+)](i) rise was abolished by removing extracellular Ca(2+), and attenuated by Trolox and a transient receptor potential M2 (TRPM2) channel blocker, N-(p-amylcinnamoyl) anthranilic acid. Other effects included mitochondrial depolarization (rhodamine-123) and increased mitochondrial reactive oxygen species (ROS) production (MitoSox), which was also abolished by Trolox. A low concentration of rotenone (5 nm) that, by itself, did not evoke a [Ca(2+)](i) rise resulted in a large (46.6 +/- 25.3 nm) Ca(2+) response when baseline [Ca(2+)](i) was increased by a 'priming' protocol that activated voltage-gated Ca(2+) channels. There was also a positive correlation between 'naturally' occurring variations in baseline [Ca(2+)](i) and the rotenone-induced [Ca(2+)](i) rise. This correlation was not seen in non-dopaminergic neurons of the substantia nigra pars reticulata (SNr). Our results show that mitochondrial ROS production is a key element in the effect of rotenone on ATP-gated K(+) channels and TRPM2-like channels in SNc neurons, and demonstrate, in these neurons (but not in the SNr), a large potentiation of rotenone-induced [Ca(2+)](i) rise by a small increase in baseline [Ca(2+)](i).


Subject(s)
Calcium/metabolism , Dopamine/metabolism , Insecticides/pharmacology , Neurons/drug effects , Reactive Oxygen Species/metabolism , Rotenone/pharmacology , Substantia Nigra/cytology , Adenosine Triphosphate/pharmacology , Analysis of Variance , Animals , Animals, Newborn , Antioxidants/pharmacology , Biophysical Phenomena/drug effects , Biophysics , Chromans/pharmacology , Cinnamates/pharmacology , Clusterin/antagonists & inhibitors , Dose-Response Relationship, Drug , Drug Interactions , Electric Stimulation , Female , Flufenamic Acid/pharmacology , Homeostasis/drug effects , In Vitro Techniques , Male , Membrane Potential, Mitochondrial/drug effects , Membrane Potentials/drug effects , Membrane Potentials/physiology , Neurons/metabolism , Patch-Clamp Techniques/methods , Rats , Rats, Wistar , Sodium/metabolism , Substantia Nigra/drug effects , Tolbutamide/pharmacology , ortho-Aminobenzoates/pharmacology
13.
J Neurosci Res ; 86(9): 2071-82, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18335520

ABSTRACT

Cross-linking of alpha-synuclein and Lewy body formation have been implicated in the dopaminergic neuronal cell death observed in Parkinson's disease (PD); the mechanisms responsible, however, are not clear. Reactive oxygen species and advanced glycation end products (AGEs) have been found in the intracellular, alpha-synuclein-positive Lewy bodies in the brains of both PD as well as incidental Lewy body disease patients, suggesting a role for AGEs in alpha-synuclein cross-linking and Lewy body formation. The aims of the present study were to determine 1) whether AGEs can induce cross-linking of alpha-synuclein peptides, 2) the progressive and time-dependent intracellular accumulation of AGEs and inclusion body formation, and 3) the effects of extracellular or exogenous AGEs on intracellular inclusion formation. We first investigated the time-dependent cross-linking of recombinant human alpha-synuclein in the presence of AGEs in vitro, then used a cell culture model based on chronic rotenone treatment of human dopaminergic neuroblastoma cells (SH-SY5Y) over a period of 1-4 weeks, in the presence of different doses of AGEs. Cells (grown on coverslips) and cell lysates, collected at the end of every week, were analyzed for the presence of intracellular reactive oxygen species, AGEs, alpha-synuclein proteins, and intracellular alpha-synuclein- and AGE-positive inclusion bodies by using immunocytochemical, biochemical, and Western blot techniques. Our results show that AGEs promote in vitro cross-linking of alpha-synuclein, that intracellular accumulation of AGEs precedes alpha-synuclein-positive inclusion body formation, and that extracellular AGEs accelerate the process of intracellular alpha-synuclein-positive inclusion body formation.


Subject(s)
Cross-Linking Reagents/pharmacology , Glycation End Products, Advanced/pharmacology , Inclusion Bodies/physiology , alpha-Synuclein/metabolism , Cell Line, Tumor , Glycation End Products, Advanced/metabolism , Humans , Inclusion Bodies/drug effects , Neuroblastoma , Reactive Oxygen Species/metabolism , Recombinant Proteins/drug effects , Recombinant Proteins/metabolism , Rotenone/pharmacology , alpha-Synuclein/drug effects
14.
Cell Biol Int ; 32(12): 1578-83, 2008 Dec.
Article in English | MEDLINE | ID: mdl-18948214

ABSTRACT

A number of chronic diseases, including neurodegenerative, cardiovascular and metabolic disorders, are associated with genetic susceptibility. Some may originate on exposure to an environmental stimulus. Regardless of genetic predisposition or external stimulus, these chronic diseases, once triggered, share an inflammatory component making them effectively persistent "wounds". There is also increasing evidence that the presence of one disease can cause activation of another apparently unrelated disease, leading to multiple disorders via activation of an immune response that 'fast forwards' disease progression. Here we review common aspects of a number of chronic disease conditions, and put forward the proposal that gap junction modulation may provide an opportunity to break the inflammatory cycle that sustains and links these disorders.


Subject(s)
Chronic Disease/drug therapy , Gap Junctions/physiology , Inflammation/drug therapy , Animals , Blood-Brain Barrier/immunology , Blood-Brain Barrier/physiopathology , Cytokines/blood , Cytokines/immunology , Endothelial Cells/immunology , Endothelial Cells/pathology , Gap Junctions/drug effects , Humans , Immune System/drug effects , Immune System/physiopathology , Inflammation/immunology , Inflammation/physiopathology , Microcirculation/immunology , Vascular Diseases/immunology , Vascular Diseases/physiopathology
15.
J Clin Neurosci ; 15(11): 1253-63, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18824359

ABSTRACT

Using a model of optic nerve ischaemia, this study investigated oxygen-glucose deprivation (OGD) on isolated rat optic nerve segments cultured in vitro. Thereafter, the effect of antisense oligodeoxynucleotides (ASODN) specific to the gap junction protein connexin43 (Cx43) was evaluated in this same model. Following exposure to OGD for 2 hours, optic nerves were maintained in interphase organotypic culture with and without exposure to Cx43 ASODN. Optic nerves were sectioned at 2 hours, 6 hours, and at days 1, 2, 3 and 6 following culture. Cell death was quantified using propidium iodide (PI) staining and specific markers for Cx43, capillaries (von Willebrand factor), astrocytes (glial fibrillary acidic protein), microglia and endothelial cells (isolectin B4) were used to evaluate these parameters in conjunction with digital light and confocal microscopy. In this model, up-regulation of Cx43 was seen at 2 hours following exposure of the optic nerve to OGD and peaked at day 3. Cx43 ASODN treatment dampened this up-regulation. Additionally, more PI labeled cells were found in the centre of control optic nerve segments than in treated nerves (p<0.01). Controls also showed evidence of capillary breakdown and increased numbers of astrocytes and activated microglia compared to Cx43 ASODN treated nerves (p<0.05). Thus, the application of Cx43 ASODN to post-ischaemic optic nerve segments significantly reduced the up-regulation of Cx43 and, subsequently, the spread of injury and a resultant inflammatory response. Cx43 up-regulation may play an important role in optic nerve injury, offering a potential avenue for treatment in optic neuropathy.


Subject(s)
Connexin 43/metabolism , Inflammation/drug therapy , Inflammation/etiology , Oligodeoxyribonucleotides, Antisense/therapeutic use , Optic Neuropathy, Ischemic/complications , Animals , Animals, Newborn , Blood Vessels/drug effects , Blood Vessels/metabolism , Cell Death/drug effects , Connexin 43/genetics , Dose-Response Relationship, Drug , Edema/drug therapy , Edema/etiology , Glial Fibrillary Acidic Protein/metabolism , Glycoproteins/metabolism , Lectins/metabolism , Oligodeoxyribonucleotides, Antisense/pharmacology , Organ Culture Techniques , Propidium , Rats , Time Factors , Versicans
16.
Neuroscience ; 380: 146-151, 2018 06 01.
Article in English | MEDLINE | ID: mdl-29625216

ABSTRACT

Receptor for advanced glycation end products (RAGE) is a multi-ligand receptor involved in the pathology of several progressive neurodegenerative disorders including Huntington's disease (HD). We previously showed that the expression of RAGE and its colocalization with ligands were increased in the striatum of HD patients, increasing with grade severity, and that the pattern of RAGE expression coincided with the medio-lateral pattern of neurodegeneration. However, the exact role of RAGE in HD remains elusive. In order to address the necessity for a direct functional study, we aimed to characterize the pattern of RAGE expression in the transgenic rat model of HD (tgHD rats). Our results showed that RAGE expression was expanded laterally in tgHD rat caudate-putamen (CPu) compared to wildtype littermates, but the expression was unchanged by disease severity. The rostro-caudal location did not affect RAGE expression. RAGE was predominantly expressed in the medium spiny neurons (MSN) where it colocalized most extensively with N-carboxymethyllysine (CML), which largely contradicts with observations from human HD brains. Overall, the tgHD rat model only partially recapitulated the pattern in striatal RAGE expression in human brains, raising a question about its reliability as an animal model for future functional studies.


Subject(s)
Corpus Striatum/metabolism , Disease Models, Animal , Huntington Disease , Neurons/metabolism , Receptor for Advanced Glycation End Products/biosynthesis , Animals , Female , Rats , Rats, Transgenic
17.
Exp Neurol ; 300: 1-12, 2018 02.
Article in English | MEDLINE | ID: mdl-29055716

ABSTRACT

Connexin43 (Cx43) hemichannels in spinal cord astrocytes are implicated in the maintenance of neuropathic pain following peripheral nerve injury. Peptide5 is a Cx43 mimetic peptide that blocks hemichannels. In this study, we investigated the effects of spinal delivery of Peptide5 on mechanical pain hypersensitivity in two mouse models of neuropathic pain, peripheral nerve injury and chemotherapy-induced peripheral neuropathy (CIPN). We demonstrated that 10days following a chronic constriction injury (CCI) of the sciatic nerve, Cx43 expression, co-localised predominantly with astrocytes, was increased in the ipsilateral L3-L5 lumbar spinal cord. An intrathecal injection of Peptide5 into nerve-injured mice, on day 10 when pain was well-established, caused significant improvement in mechanical pain hypersensitivity 8h after injection. Peptide5 treatment resulted in significantly reduced Cx43, and microglial and astrocyte activity in the dorsal horn of the spinal cord, as compared to control saline-treated CCI mice. Further in vitro investigations on primary astrocyte cultures showed that 1h pre-treatment with Peptide5 significantly reduced adenosine triphosphate (ATP) release in response to extracellular calcium depletion. Since ATP is a known activator of the NOD-like receptor protein 3 (NLRP3) inflammasome complex, a key mediator of neuroinflammation, we examined the effects of Peptide5 treatment on NLRP3 inflammasome expression. We found that NLRP3, its adaptor apoptosis-associated spec-like protein (ASC) and caspase-1 protein were increased in the ipsilateral spinal cord of CCI mice and reduced to naïve levels following Peptide5 treatment. In the models of oxaliplatin- and paclitaxel-induced peripheral neuropathy, treatment with Peptide5 had no effect on mechanical pain hypersensitivity. Interestingly, in these CIPN models, although spinal Cx43 expression was significantly increased at day 13 following chemotherapy, NLRP3 expression was not altered. These results suggest that the analgesic effect of Peptide5 is specifically achieved by reducing NLRP3 expression. Together, our findings demonstrate that blocking Cx43 hemichannels with Peptide5 after nerve injury attenuates mechanical pain hypersensitivity by specifically targeting the NLRP3 inflammasome in the spinal cord.


Subject(s)
Biomimetic Materials/administration & dosage , Connexin 43/administration & dosage , Hyperalgesia/drug therapy , NLR Family, Pyrin Domain-Containing 3 Protein/antagonists & inhibitors , Neuralgia/drug therapy , Peptide Fragments/administration & dosage , Animals , Hyperalgesia/metabolism , Inflammasomes/antagonists & inhibitors , Inflammasomes/metabolism , Injections, Spinal , Male , Mice , Mice, Inbred C57BL , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Neuralgia/metabolism , Treatment Outcome
18.
Tissue Cell ; 39(1): 35-46, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17316726

ABSTRACT

The cellular distributions of the growth factors FGF-2 and VEGF, and their receptors FGFR1, FGFR2 and FGFR3, and VEGFR-2 respectively, were visualized by immunohistochemistry and light microscopy in sections of growing red deer antler. Both of these signalling systems were widely expressed in the integument and osteocartilaginous compartments. FGF-2 was found in the same cells as all three FGFRs, indicating that FGF signalling may be principally autocrine. The patterns of labelling for VEGF and its receptor were similar to those seen for FGF-2 and FGFR-3, in both compartments. Our data are consistent with the findings of others in suggesting that FGF-2 induces expression of VEGF, to stimulate and maintain high rates of neovascularisation and angiogenesis, thereby providing nutrients to both velvet and bone as they rapidly grow and develop. The presence of FGF and VEGF and their receptors in epithelial cells suggests that these signalling systems play a role in skin development, raising the possibility that one or both may be involved in the close coupling of the coordinated growth of the integument and osteocartilage of antler, a process which is poorly understood at present.


Subject(s)
Antlers/growth & development , Antlers/metabolism , Deer/physiology , Fibroblast Growth Factor 2/metabolism , Receptors, Growth Factor/metabolism , Vascular Endothelial Growth Factor A/metabolism , Animals , Antlers/blood supply , Blood Vessels/growth & development , Blood Vessels/metabolism , Bone Development/physiology , Deer/anatomy & histology , Immunohistochemistry , Male , Neovascularization, Physiologic/physiology , Receptor, Fibroblast Growth Factor, Type 1/metabolism , Receptor, Fibroblast Growth Factor, Type 2/metabolism , Receptor, Fibroblast Growth Factor, Type 3/metabolism , Signal Transduction/physiology , Skin/growth & development , Vascular Endothelial Growth Factor Receptor-2/metabolism
19.
Neuroscientist ; 23(1): 7-15, 2017 Feb.
Article in English | MEDLINE | ID: mdl-26290448

ABSTRACT

Engaging young people with science is essential to ensuring a scientifically literate society. Furthermore, it is important to enable access to a variety of sciences during adolescence, when individuals are making decisions about their future educational and career paths. The Brain Bee Challenge (BBC) is a quiz-based international neuroscience outreach program for high school students. We wished to determine what influence exposure to the scientific research environment had on the highest achievers' later choices in education, their career expectations, and their perspectives toward science. Semistructured interviews were carried out with seven of the past winners of the New Zealand National BBC finals. Analysis involved thematic coding to investigate the impact of BBC involvement and potential longer term consequences. Second-order coding found critical themes identified by participants. These themes highlight the value of research institution-led outreach activities that extend high achievers beyond the school curriculum. In addition to subject-specific influences, there were multiple benefits acknowledged at a personal or individual level, including socialization and identity development, further demonstrating the importance of such engagement activities.

20.
J Neurotrauma ; 34(3): 707-719, 2017 02.
Article in English | MEDLINE | ID: mdl-27629792

ABSTRACT

Blocking of Connexin43 hemichannels, the main gap junction protein located on astrocytes in the central nervous system, has been shown to reduce neural injury in a number of models. We demonstrated previously that local administration of a Connexin43 mimetic peptide, Peptide5, reduces secondary tissue damage after spinal cord injury (SCI). Here, we investigated whether acute systemic delivery of Peptide5 is also protective in a model of SCI. Rats were subjected to a mild spinal cord contusion using the Multicentre Animal Spinal Cord Injury Study impactor and were injected intraperitoneally with Peptide5 or a scrambled peptide immediately and at 2 h and 4 h post-injury. Rats were tested for locomotor recovery and pain hypersensitivity and euthanized at 8 h, 24 h, two weeks, or six weeks post-injury. Compared with control rats, Peptide5 treated rats showed significant improvement in hindlimb locomotor function between three and six weeks post-injury and reductions in at-level mechanical allodynia at weeks one and six post-injury. Immunohistochemistry showed that Peptide5 treatment led to a reduction in total Connexin43 and increased phosphorylated Connexin43 at 8 h compared with scrambled peptide. At two and six weeks, lesion size, the astrocytic and the activated macrophage, and/or microglial response were all decreased in the Peptide5 animals. In addition, neuronal cell numbers were higher in the Peptide5 animals compared with the scrambled peptide treated rats at two and six weeks. These results show for the first time that systemic administration of Peptide5 to block the pathological opening of Connexin43 hemichannels is a feasible treatment strategy in this setting, ameliorating the secondary SCI.


Subject(s)
Biomimetic Materials/administration & dosage , Connexin 43/administration & dosage , Recovery of Function/drug effects , Recovery of Function/physiology , Spinal Cord Injuries/drug therapy , Age Factors , Animals , Cell Survival/drug effects , Cell Survival/physiology , Drug Administration Routes , Female , Neurons/drug effects , Neurons/pathology , Rats , Rats, Sprague-Dawley , Spinal Cord Injuries/pathology
SELECTION OF CITATIONS
SEARCH DETAIL