102 results on '"Dev KK"'
Search Results
2. The Effect of Donepezil Hydrochloride in the Twitcher Mouse Model of Krabbe Disease.
- Author
-
Papakyriakopoulou P, Valsami G, and Dev KK
- Subjects
- Animals, Mice, Inbred C57BL, Mice, Cholinesterase Inhibitors pharmacology, Cholinesterase Inhibitors therapeutic use, Brain metabolism, Brain drug effects, Brain pathology, Neuroglia drug effects, Neuroglia metabolism, Neuroglia pathology, Behavior, Animal drug effects, Donepezil pharmacology, Donepezil therapeutic use, Disease Models, Animal, Leukodystrophy, Globoid Cell drug therapy, Leukodystrophy, Globoid Cell pathology, Myelin Sheath metabolism, Myelin Sheath drug effects
- Abstract
Krabbe disease (KD) is a rare demyelinating disorder characterized by demyelination caused by mutations in the GALC gene, resulting in toxic accumulation of psychosine. Psychosine has been identified as detrimental to oligodendrocytes, leading to demyelination through diverse hypothesized pathways. Reducing demyelination is essential to maintain neurological function in KD; however, therapeutic interventions are currently limited. Acetylcholinesterase inhibitors (AChEi) are commonly used for symptomatic management of Alzheimer's Disease and are suggested to have potential disease-modifying effects, including regulating myelin state. In particular, donepezil, an AChEi, has demonstrated promising effects in cellular and animal models, including promotion of the expression of myelin-related genes and reduction of glial cell reactivity. This drug also acts as an agonist for sigma-1 receptors (Sig-1R), which are implicated in demyelination diseases. In the context of drug repurposing, here, we demonstrate that administration of donepezil has protective effects in the twitcher mouse model of KD. We provide data showing that donepezil preserves myelin and reduces glial cell reactivity in the brains of twitcher mice. Moreover, donepezil also improves behavioral phenotypes and increases lifespan in twitcher animals. These findings suggest that donepezil, with its dual activity as an AChE inhibitor and Sig-1R agonist, may hold promise as a therapeutic candidate for demyelinating diseases, including KD., (© 2024. The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
3. Pharmacokinetic Study of Fingolimod Nasal Films Administered via Nose-to-Brain Route in C57BL/6 J Mice as Potential Treatment for Multiple Sclerosis.
- Author
-
Papakyriakopoulou P, Balafas E, Kostomitsopoulos N, Rekkas DM, Dev KK, and Valsami G
- Subjects
- Animals, Mice, Biological Availability, Immunosuppressive Agents pharmacokinetics, Immunosuppressive Agents administration & dosage, Nasal Mucosa metabolism, Drug Delivery Systems, Administration, Oral, Male, Female, Fingolimod Hydrochloride pharmacokinetics, Fingolimod Hydrochloride administration & dosage, Administration, Intranasal, Mice, Inbred C57BL, Multiple Sclerosis drug therapy, Multiple Sclerosis metabolism, Brain metabolism
- Abstract
Background: Fingolimod hydrochloride (FH) has emerged as a vital medication for managing Multiple Sclerosis (MS). Despite its high oral bioavailability of 93%, it is plagued by slow oral absorption (T
max = 8-12 h) and extensive hepatic metabolism. Intranasal administration has emerged as an alternative to address these limitations, ensuring efficient central nervous system delivery and minimizing peripheral exposure and first-pass metabolism., Objective: This study aims to develop and evaluate FH nasal films for enhanced drug delivery., Methods: A Design of Experiments approach was employed to formulate FH nasal films, utilizing HPMC E50 as a film-forming polymer, PEG 400 as a plasticizer, and Me-β-CD as a permeation enhancer. Two formulations with superior in vitro and ex vivo performance were selected for in vivo evaluation. A comparative pharmacokinetic study was conducted in C57BL/6 J mice in the brain and serum after administration of nasal films and oral FH solution, respectively. Sparse sampling and non-compartmental analysis were used., Results: FH nasal films efficiently delivered the drug to both serum (Cmax(F3) = 0.35 ± 0.021, Cmax(F4) = 0.38 ± 0.029 μg/mL) and brain (Cmax(F3) = 0.39 ± 0.05, Cmax(F4) = 0.44 ± 0.048 μg/mL), achieving higher levels than oral delivery. Brain relative bioavailability (% Frel (0-6 h) ) was 519% and 534%, while serum % Frel (0-6 h) was 295% and 343%., Conclusions: The rapid nose-to-brain delivery within 30 min, in contrast to 10-h Tmax of the oral solution, indicates the potential of a combined IN and oral treatment regimen. This approach could expedite the attainment of steady-state concentrations, offering a promising method for managing multiple sclerosis (MS)., (© 2024. The Author(s), under exclusive licence to Springer Science+Business Media, LLC, part of Springer Nature.)- Published
- 2024
- Full Text
- View/download PDF
4. The Effects of the S1P Receptor Agonist Fingolimod (FTY720) on Central and Peripheral Myelin in Twitcher Mice.
- Author
-
Béchet S and Dev KK
- Abstract
Krabbe's disease (KD) is caused by mutations in the lysosomal enzyme galactocerebrosidase and is associated with psychosine toxicity. The sphingosine 1-phosphate receptor (S1PR) agonist fingolimod (FTY720) attenuates psychosine-induced cell death of human astrocytes, demyelination in cerebellar slices, as well as demyelination in the central nervous system of twitcher mice. Psychosine also accumulates in the peripheral nervous system in twitcher mice; however, effects of fingolimod on this peripheral myelin have not been examined. The aim of this study was to investigate the effects of fingolimod administration on peripheral and central markers of myelination. Here, we report that fingolimod administration (1 mg/kg/day) from postnatal day 5 (PND) onwards did not alter peripheral demyelination in the sciatic nerve of twitcher mice, despite significantly reducing myelin debris, glial reactivity, and neuronal damage in the cerebellum. We also find fingolimod administration improves twitching and mobility scores in twitcher mice. Importantly, we find that fingolimod significantly increases the lifespan of twitcher mice by approximately 5 days. These findings suggest differential effects of fingolimod on peripheral and central neuropathy in twitcher mice, which may explain its modest efficacy on behavior and lifespan.
- Published
- 2024
- Full Text
- View/download PDF
5. Elucidating the Therapeutic Utility of Olaparib in Sulfatide-Induced Human Astrocyte Toxicity and Neuroinflammation.
- Author
-
Mekhaeil M, Conroy MJ, and Dev KK
- Subjects
- Humans, Astrocytes, Neuroinflammatory Diseases, Poly(ADP-ribose) Polymerase Inhibitors toxicity, Reactive Oxygen Species, Sulfoglycosphingolipids, Leukodystrophy, Metachromatic genetics, Leukodystrophy, Metachromatic therapy
- Abstract
Metachromatic leukodystrophy (MLD) is a severe demyelinating, autosomal recessive genetic leukodystrophy, with no curative treatment. The disease is underpinned by mutations in the arylsulfatase A gene (ARSA), resulting in deficient activity of this lysosomal enzyme, and consequential accumulation of galactosylceramide-3-O-sulfate (sulfatide) in the brain. Most of the effects in the brain have been attributed to the accumulation of sulfatides in oligodendrocytes and their cell damage. In contrast, less is known regarding sulfatide toxicity in astrocytes. Poly (ADP-ribose) polymerase (PARP) inhibitors are anti-cancer therapeutics that have proven efficacy in preclinical models of many neurodegenerative and inflammatory diseases, but have never been tested for MLD. Here, we examined the toxic effect of sulfatides on human astrocytes and restoration of this cell damage by the marketed PARP-1 inhibitor, Olaparib. Cultured human astrocytes were treated with increasing concentrations of sulfatides (5-100 μM) with or without Olaparib (100 nM). Cell viability assays were used to ascertain whether sulfatide-induced toxicity was rescued by Olaparib. Immunofluorescence, calcium (Ca
2+ ) imaging, ROS, and mitochondrial damage assays were also used to explore the effects of sulfatides and Olaparib. ELISAs were performed and chemotaxis of peripheral blood immune cells was measured to examine the effects of Olaparib on sulfatide-induced inflammation in human astrocytes. Here, we established a concentration-dependent (EC50 ∼20 μM at 24 h) model of sulfatide-induced astrocyte toxicity. Our data demonstrate that sulfatide-induced astrocyte toxicity involves (i) PARP-1 activation, (ii) pro-inflammatory cytokine release, and (iii) enhanced chemoattraction of peripheral blood immune cells. Moreover, these sulfatide-induced effects were attenuated by Olaparib (IC50 ∼100 nM). In addition, sulfatide caused impairments of ROS production, mitochondrial stress, and Ca2+ signaling in human astrocytes, that were indicative of metabolic alterations and that were also alleviated by Olaparib (100 nM) treatment. Our data support the hypothesis that sulfatides can drive astrocyte cell death and demonstrate that Olaparib can dampen many facets of sulfatide-induced toxicity, including, mitochondrial stress, inflammatory responses, and communication between human astrocytes and peripheral blood immune cells. These data are suggestive of potential therapeutic utility of PARP inhibitors in the sphere of rare demyelinating diseases, and in particular MLD. Graphical abstract. Proposed mechanism of action of Olaparib in sulfatide-treated astrocytes. Human astrocytes treated for 24 h with sulfatides increase PARP-1 expression and die. PARP-1 overexpression is modulated by Ca2+ release from the endoplasmic reticulum, thus enhancing intracellular Ca2+ concentration. PARP-1 inhibition with Olaparib reduces Ca2+ influx and cell death. Olaparib also decreases IL-6, IL-8, IL-17, and CX3CL1 release from sulfatide-stimulated astrocytes, suggesting that PARP-1 plays a role in dampening neuroinflammation in MLD. This is confirmed by the reduction of immune cell migration such as lymphocytes, NK cells, and T cells towards sulfatide-treated astrocytes. Moreover, mitochondrial stress and ROS production induced by sulfatides are rescued by PARP-1 inhibition. Future studies will focus on the signaling cascades triggered by PARP-1-mediated currents in reactive astrocytes and Olaparib as a potential therapeutic target for MLD., (© 2023. The Author(s).)- Published
- 2023
- Full Text
- View/download PDF
6. Olaparib Attenuates Demyelination and Neuroinflammation in an Organotypic Slice Culture Model of Metachromatic Leukodystrophy.
- Author
-
Mekhaeil M, Conroy MJ, and Dev KK
- Subjects
- Animals, Mice, Cerebroside-Sulfatase genetics, Cerebroside-Sulfatase metabolism, Sulfoglycosphingolipids metabolism, Neuroinflammatory Diseases, Poly(ADP-ribose) Polymerase Inhibitors, Leukodystrophy, Metachromatic genetics, Leukodystrophy, Metachromatic metabolism, Demyelinating Diseases
- Abstract
Metachromatic leukodystrophy (MLD) is a severe demyelinating, autosomal recessive genetic leukodystrophy. The disease is underpinned by mutations in the arylsulfatase A gene (ARSA), resulting in deficient activity of the arylsulfatase A lysosomal enzyme and consequential accumulation of galactosylceramide-3-O-sulfate (sulfatide) in the brain. Using an ex vivo murine-derived organotypic cerebellar slice culture model, we demonstrate that sulfatide induces demyelination in a concentration-dependent manner. Interestingly, our novel data demonstrate that sulfatide-induced demyelination is underpinned by PARP-1 activation, oligodendrocyte loss, pro-inflammatory cytokine expression, astrogliosis, and microgliosis. Moreover, such sulfatide-induced effects can be attenuated by the treatment with the poly (ADP-ribose) polymerase 1 (PARP-1) inhibitor Olaparib (IC50∼100 nM) suggesting that this small molecule may be neuroprotective and limit toxin-induced demyelination. Our data support the idea that sulfatide is a key driver of demyelination and neuroinflammation in MLD and suggest that PARP-1 inhibitors have therapeutic utility in the sphere of rare demyelinating disease., (© 2023. The Author(s).)
- Published
- 2023
- Full Text
- View/download PDF
7. The Effects of Antipsychotics in Experimental Models of Krabbe Disease.
- Author
-
Sharma K and Dev KK
- Abstract
The role of altered myelin in the onset and development of schizophrenia and changes in myelin due to antipsychotics remains unclear. Antipsychotics are D
2 receptor antagonists, yet D2 receptor agonists increase oligodendrocyte progenitor numbers and limit oligodendrocyte injury. Conflicting studies suggest these drugs promote the differentiation of neural progenitors to oligodendrocyte lineage, while others report antipsychotics inhibit the proliferation and differentiation of oligodendrocyte precursors. Here, we utilised in-vitro (human astrocytes), ex-vivo (organotypic slice cultures) and in-vivo (twitcher mouse model) experimental study designs of psychosine-induced demyelination, a toxin that accumulates in Krabbe disease (KD), to investigate direct effects of antipsychotics on glial cell dysfunction and demyelination. Typical and atypical antipsychotics, and selective D2 and 5HT2A receptor antagonists, attenuated psychosine-induced cell viability, toxicity, and morphological aberrations in human astrocyte cultures. Haloperidol and clozapine reduced psychosine-induced demyelination in mouse organotypic cerebellar slices. These drugs also attenuated the effects of psychosine on astrocytes and microglia and restored non-phosphorylated neurofilament levels, indicating neuroprotective effects. In the demyelinating twitcher mouse model of KD, haloperidol improved mobility and significantly increased the survival of these animals. Overall, this study suggests that antipsychotics directly regulate glial cell dysfunction and exert a protective effect on myelin loss. This work also points toward the potential use of these pharmacological agents in KD.- Published
- 2023
- Full Text
- View/download PDF
8. Investigating the Effects of Olaparib on the Susceptibility of Glioblastoma Multiforme Tumour Cells to Natural Killer Cell-Mediated Responses.
- Author
-
Moran J, Mylod E, Kane LE, Marion C, Keenan E, Mekhaeil M, Lysaght J, Dev KK, O'Sullivan J, and Conroy MJ
- Abstract
Glioblastoma multiforme (GBM) is the most common adult primary brain malignancy, with dismal survival rates of ~14.6 months. The current standard-of-care consists of surgical resection and chemoradiotherapy, however the treatment response is limited by factors such as tumour heterogeneity, treatment resistance, the blood-brain barrier, and immunosuppression. Several immunotherapies have undergone clinical development for GBM but demonstrated inadequate efficacy, yet future combinatorial approaches are likely to hold more promise. Olaparib is FDA-approved for BRCA-mutated advanced ovarian and breast cancer, and clinical studies have revealed its utility as a safe and efficacious radio- and chemo-sensitiser in GBM. The ability of Olaparib to enhance natural killer (NK) cell-mediated responses has been reported in prostate, breast, and lung cancer. This study examined its potential combination with NK cell therapies in GBM by firstly investigating the susceptibility of the GBM cell line T98G to NK cells and, secondly, examining whether Olaparib can sensitise T98G cells to NK cell-mediated responses. Here, we characterise the NK receptor ligand profile of T98G cells and demonstrate that Olaparib does not dampen T98G susceptibility to NK cells or elicit immunomodulatory effects on the function of NK cells. This study provides novel insights into the potential combination of Olaparib with NK cell therapies for GBM.
- Published
- 2023
- Full Text
- View/download PDF
9. Fingolimod Rescues Memory and Improves Pathological Hallmarks in the 3xTg-AD Model of Alzheimer's Disease.
- Author
-
Fagan SG, Bechet S, and Dev KK
- Subjects
- Amyloid beta-Peptides, Amyloid beta-Protein Precursor, Animals, Disease Models, Animal, Fingolimod Hydrochloride pharmacology, Fingolimod Hydrochloride therapeutic use, Mice, Mice, Transgenic, tau Proteins, Alzheimer Disease pathology
- Abstract
Therapeutic strategies for Alzheimer's disease (AD) have largely focused on the regulation of amyloid pathology while those targeting tau pathology, and inflammatory mechanisms are less explored. In this regard, drugs with multimodal and concurrent targeting of Aβ, tau, and inflammatory processes may offer advantages. Here, we investigate one such candidate drug in the triple transgenic 3xTg-AD mouse model of AD, namely the disease-modifying oral neuroimmunomodulatory therapeutic used in patients with multiple sclerosis, called fingolimod. In this study, administration of fingolimod was initiated after behavioral symptoms are known to emerge, at 6 months of age. Treatment continued to 12 months when behavioral tests were performed and thereafter histological and biochemical analysis was conducted on postmortem tissue. The results demonstrate that fingolimod reverses deficits in spatial working memory at 8 and 12 months of age as measured by novel object location and Morris water maze tests. Inflammation in the brain is alleviated as demonstrated by reduced Iba1-positive and CD3-positive cell number, less ramified microglial morphology, and improved cytokine profile. Finally, treatment with fingolimod was shown to reduce phosphorylated tau and APP levels in the hippocampus and cortex. These results highlight the potential of fingolimod as a multimodal therapeutic for the treatment of AD., (© 2022. The Author(s).)
- Published
- 2022
- Full Text
- View/download PDF
10. Existing Evidence for the Repurposing of PARP-1 Inhibitors in Rare Demyelinating Diseases.
- Author
-
Mekhaeil M, Dev KK, and Conroy MJ
- Abstract
Over the past decade, Poly (ADP-ribose) polymerase-1 (PARP-1) inhibitors have arisen as a novel and promising targeted therapy for breast cancer gene (BRCA)-mutated ovarian and breast cancer patients. Therapies targeting the enzyme, PARP-1, have since established their place as maintenance drugs for cancer. Here, we present existing evidence that implicates PARP-1 as a player in the development and progression of both malignancy and demyelinating disease. These findings, together with the proven clinical efficacy and marketed success of PARP-1 inhibitors in cancer, present the repurposing of these drugs for demyelinating diseases as a desirable therapeutic concept. Indeed, PARP-1 inhibitors are noted to demonstrate neuroprotective effects in demyelinating disorders such as multiple sclerosis and Parkinson's disease, further supporting the use of these drugs in demyelinating, neuroinflammatory, and neurodegenerative diseases. In this review, we discuss the potential for repurposing PARP-1 inhibitors, with a focus on rare demyelinating diseases. In particular, we address the possible use of PARP-1 inhibitors in examples of rare leukodystrophies, for which there are a paucity of treatment options and an urgent need for novel therapeutic approaches.
- Published
- 2022
- Full Text
- View/download PDF
11. Hybrid Nanoparticles as a Novel Tool for Regulating Psychosine-Induced Neuroinflammation and Demyelination In Vitro and Ex vivo.
- Author
-
Clementino A, Velasco-Estevez M, Buttini F, Sonvico F, and Dev KK
- Subjects
- Animals, Mice, Neuroinflammatory Diseases, Psychosine pharmacology, Psychosine therapeutic use, Demyelinating Diseases chemically induced, Demyelinating Diseases drug therapy, Leukodystrophy, Globoid Cell drug therapy, Nanoparticles
- Abstract
Polymeric nanoparticles are being extensively investigated as an approach for brain delivery of drugs, especially for their controlled release and targeting capacity. Nose-to-brain administration of nanoparticles, bypassing the blood brain barrier, offers a promising strategy to deliver drugs to the central nervous system. Here, we investigated the potential of hybrid nanoparticles as a therapeutic approach for demyelinating diseases, more specifically for Krabbe's disease. This rare leukodystrophy is characterized by the lack of enzyme galactosylceramidase, leading to the accumulation of toxic psychosine in glial cells causing neuroinflammation, extensive demyelination and death. We present evidence that lecithin/chitosan nanoparticles prevent damage associated with psychosine by sequestering the neurotoxic sphingolipid via physicochemical hydrophobic interactions. We showed how nanoparticles prevented the cytotoxicity caused by psychosine in cultured human astrocytes in vitro, and how the nanoparticle size and PDI augmented while the electrostatic charges of the surface decreased, suggesting a direct interaction between psychosine and the nanoparticles. Moreover, we studied the effects of nanoparticles ex vivo using mouse cerebellar organotypic cultures, observing that nanoparticles prevented the demyelination and axonal damage caused by psychosine, as well as a moderate prevention of the astrocytic death. Taken together, these results suggest that lecithin-chitosan nanoparticles are a potential novel delivery system for drugs for certain demyelinating conditions such as Krabbe's disease, due to their dual effect: not only are they an efficient platform for drug delivery, but they exert a protective effect themselves in tampering the levels of psychosine accumulation., (© 2021. The Author(s).)
- Published
- 2021
- Full Text
- View/download PDF
12. EBI2 Is Temporarily Upregulated in MO3.13 Oligodendrocytes during Maturation and Regulates Remyelination in the Organotypic Cerebellar Slice Model.
- Author
-
Velasco-Estevez M, Koch N, Klejbor I, Laurent S, Dev KK, Szutowicz A, Sailer AW, and Rutkowska A
- Subjects
- Animals, Brain metabolism, Cerebellum metabolism, Mice, Mice, Inbred C57BL, Mice, Knockout, Oligodendroglia metabolism, Receptors, G-Protein-Coupled genetics, Remyelination, Stem Cells metabolism, Brain cytology, Cerebellum cytology, Oligodendroglia cytology, Receptors, G-Protein-Coupled metabolism, Stem Cells cytology
- Abstract
The EBI2 receptor regulates the immune system and is expressed in various immune cells including B and T lymphocytes. It is also expressed in astrocytes in the central nervous system (CNS) where it regulates pro-inflammatory cytokine release, cell migration and protects from chemically induced demyelination. Its signaling and expression are implicated in various diseases including multiple sclerosis, where its expression is increased in infiltrating immune cells in the white matter lesions. Here, for the first time, the EBI2 protein in the CNS cells in the human brain was examined. The function of the receptor in MO3.13 oligodendrocytes, as well as its role in remyelination in organotypic cerebellar slices, were investigated. Human brain sections were co-stained for EBI2 receptor and various markers of CNS-specific cells and the human oligodendrocyte cell line MO3.13 was used to investigate changes in EBI2 expression and cellular migration. Organotypic cerebellar slices prepared from wild-type and cholesterol 25-hydroxylase knock-out mice were used to study remyelination following lysophosphatidylcholine (LPC)-induced demyelination. The data showed that EBI2 receptor is present in OPCs but not in myelinating oligodendrocytes in the human brain and that EBI2 expression is temporarily upregulated in maturing MO3.13 oligodendrocytes. Moreover, we show that migration of MO3.13 cells is directly regulated by EBI2 and that its signaling is necessary for remyelination in cerebellar slices post-LPC-induced demyelination. The work reported here provides new information on the expression and role of EBI2 in oligodendrocytes and myelination and provides new tools for modulation of oligodendrocyte biology and therapeutic approaches for demyelinating diseases.
- Published
- 2021
- Full Text
- View/download PDF
13. Spatiotemporal immunolocalisation of REST in the brain of healthy ageing and Alzheimer's disease rats.
- Author
-
Mampay M, Velasco-Estevez M, Rolle SO, Chaney AM, Boutin H, Dev KK, Moeendarbary E, and Sheridan GK
- Subjects
- Alzheimer Disease genetics, Alzheimer Disease physiopathology, Amyloid beta-Protein Precursor genetics, Animals, Disease Models, Animal, Female, Healthy Aging physiology, Humans, Learning physiology, Male, Memory physiology, Mutation, Neurons, Presenilin-1 genetics, Rats, Rats, Inbred F344, Rats, Transgenic, Repressor Proteins metabolism, Spatio-Temporal Analysis, Synaptophysin analysis, Synaptophysin metabolism, Alzheimer Disease pathology, CA1 Region, Hippocampal pathology, Frontal Lobe pathology, Healthy Aging pathology, Repressor Proteins analysis
- Abstract
In the brain, REST (Repressor Element-1 Silencing Transcription factor) is a key regulator of neuron cell-specific gene expression. Nuclear translocation of neuronal REST has been shown to be neuroprotective in a healthy ageing context. In contrast, inability to upregulate nuclear REST is thought to leave ageing neurons vulnerable to neurodegenerative stimuli, such as Alzheimer's disease (AD) pathology. Hippocampal and cortical neurons are known to be particularly susceptible to AD-associated neurodegeneration. However, REST expression has not been extensively characterised in the healthy ageing brain. Here, we examined the spatiotemporal immunolocalisation of REST in the brains of healthy ageing wild-type Fischer-344 and transgenic Alzheimer's disease rats (TgF344-AD). Nuclear expression of REST increased from 6 months to 18 months of age in the hippocampus, frontal cortex and subiculum of wild-type rats, but not in TgF344-AD rats. No changes in REST were measured in more posterior cortical regions or in the thalamus. Interestingly, levels of the presynaptic marker synaptophysin, a known gene target of REST, were lower in CA1 hippocampal neurons of 18-month TgF344-AD rats compared to 18-month wild-types, suggesting that elevated nuclear REST may protect against synapse loss in the CA1 of 18-month wild-type rats. High REST expression in ageing wild-type rats did not, however, protect against axonal loss nor against astroglial reactivity in the hippocampus. Taken together, our data confirm that changes in nuclear REST expression are context-, age- and brain region-specific. Moreover, key brain structures involved in learning and memory display elevated REST expression in healthy ageing wild-type rats but not TgF344-AD rats., (© 2020 The Authors. FEBS Open Bio published by John Wiley & Sons Ltd on behalf of Federation of European Biochemical Societies.)
- Published
- 2021
- Full Text
- View/download PDF
14. Fingolimod Rescues Demyelination in a Mouse Model of Krabbe's Disease.
- Author
-
Béchet S, O'Sullivan SA, Yssel J, Fagan SG, and Dev KK
- Subjects
- Animals, Astrocytes metabolism, Behavior, Animal, Female, Leukodystrophy, Globoid Cell psychology, Male, Mice, Mice, Inbred C57BL, Mice, Neurologic Mutants, Microglia metabolism, Myelin Sheath metabolism, Neurofilament Proteins metabolism, Neurons metabolism, Phosphorylation, Proprotein Convertases drug effects, Purkinje Cells metabolism, Serine Endopeptidases drug effects, Demyelinating Diseases drug therapy, Fingolimod Hydrochloride therapeutic use, Leukodystrophy, Globoid Cell drug therapy
- Abstract
Krabbe's disease is an infantile neurodegenerative disease, which is affected by mutations in the lysosomal enzyme galactocerebrosidase, leading to the accumulation of its metabolite psychosine. We have shown previously that the S1P receptor agonist fingolimod (FTY720) attenuates psychosine-induced glial cell death and demyelination both in vitro and ex vivo models. These data, together with a lack of therapies for Krabbe's disease, prompted the current preclinical study examining the effects of fingolimod in twitcher mice, a murine model of Krabbe's disease. Twitcher mice, both male and female, carrying a natural mutation in the galc gene were given fingolimod via drinking water (1 mg/kg/d). The direct impact of fingolimod administration was assessed via histochemical and biochemical analysis using markers of myelin, astrocytes, microglia, neurons, globoid cells, and immune cells. The effects of fingolimod on twitching behavior and life span were also demonstrated. Our results show that treatment of twitcher mice with fingolimod significantly rescued myelin levels compared with vehicle-treated animals and also regulated astrocyte and microglial reactivity. Furthermore, nonphosphorylated neurofilament levels were decreased, indicating neuroprotective and neurorestorative processes. These protective effects of fingolimod on twitcher mice brain pathology was reflected by an increased life span of fingolimod-treated twitcher mice. These in vivo findings corroborate initial in vitro studies and highlight the potential use of S1P receptors as drug targets for treatment of Krabbe's disease. SIGNIFICANCE STATEMENT This study demonstrates that the administration of the therapy known as fingolimod in a mouse model of Krabbe's disease (namely, the twitcher mouse model) significantly rescues myelin levels. Further, the drug fingolimod also regulates the reactivity of glial cells, astrocytes and microglia, in this mouse model. These protective effects of fingolimod result in an increased life span of twitcher mice., (Copyright © 2020 Béchet et al.)
- Published
- 2020
- Full Text
- View/download PDF
15. Inhibition of Piezo1 attenuates demyelination in the central nervous system.
- Author
-
Velasco-Estevez M, Gadalla KKE, Liñan-Barba N, Cobb S, Dev KK, and Sheridan GK
- Subjects
- Animals, Astrocytes metabolism, Cell Differentiation physiology, Central Nervous System drug effects, Central Nervous System metabolism, Demyelinating Diseases metabolism, Ion Channels metabolism, Mechanotransduction, Cellular physiology, Mice, Neural Stem Cells drug effects, Neural Stem Cells metabolism, Neurogenesis drug effects, Astrocytes drug effects, Demyelinating Diseases drug therapy, Ion Channels drug effects, Peptides pharmacology
- Abstract
Piezo1 is a mechanosensitive ion channel that facilitates the translation of extracellular mechanical cues to intracellular molecular signaling cascades through a process termed, mechanotransduction. In the central nervous system (CNS), mechanically gated ion channels are important regulators of neurodevelopmental processes such as axon guidance, neural stem cell differentiation, and myelination of axons by oligodendrocytes. Here, we present evidence that pharmacologically mediated overactivation of Piezo1 channels negatively regulates CNS myelination. Moreover, we found that the peptide GsMTx4, an antagonist of mechanosensitive cation channels such as Piezo1, is neuroprotective and prevents chemically induced demyelination. In contrast, the positive modulator of Piezo1 channel opening, Yoda-1, induces demyelination and neuronal damage. Using an ex vivo murine-derived organotypic cerebellar slice culture model, we demonstrate that GsMTx4 attenuates demyelination induced by the cytotoxic lipid, psychosine. Importantly, we confirmed the potential therapeutic effects of GsMTx4 peptide in vivo by co-administering it with lysophosphatidylcholine (LPC), via stereotactic injection, into the cerebral cortex of adult mice. GsMTx4 prevented both demyelination and neuronal damage usually caused by the intracortical injection of LPC in vivo; a well-characterized model of focal demyelination. GsMTx4 also attenuated both LPC-induced astrocyte toxicity and microglial reactivity within the lesion core. Overall, our data suggest that pharmacological activation of Piezo1 channels induces demyelination and that inhibition of mechanosensitive channels, using GsMTx4, may alleviate the secondary progressive neurodegeneration often present in the latter stages of demyelinating diseases., (© 2019 Wiley Periodicals, Inc.)
- Published
- 2020
- Full Text
- View/download PDF
16. Piezo1 regulates calcium oscillations and cytokine release from astrocytes.
- Author
-
Velasco-Estevez M, Rolle SO, Mampay M, Dev KK, and Sheridan GK
- Subjects
- Animals, Astrocytes drug effects, Calcium Signaling drug effects, Cells, Cultured, Cerebral Cortex cytology, Cerebral Cortex drug effects, Cerebral Cortex metabolism, Female, Lipopolysaccharides toxicity, Male, Mice, Mice, Inbred C57BL, Astrocytes metabolism, Calcium Signaling physiology, Cytokines metabolism, Ion Channels biosynthesis
- Abstract
Astrocytes are important for information processing in the brain and they achieve this by fine-tuning neuronal communication via continuous uptake and release of biochemical modulators of neurotransmission and synaptic plasticity. Often overlooked are their important functions in mechanosensation. Indeed, astrocytes can detect pathophysiological changes in the mechanical properties of injured, ageing, or degenerating brain tissue. We have recently shown that astrocytes surrounding mechanically-stiff amyloid plaques upregulate the mechanosensitive ion channel, Piezo1. Moreover, ageing transgenic Alzheimer's rats harboring a chronic peripheral bacterial infection displayed enhanced Piezo1 expression in amyloid plaque-reactive astrocytes of the hippocampus and cerebral cortex. Here, we have shown that the bacterial endotoxin, lipopolysaccharide (LPS), also upregulates Piezo1 in primary mouse cortical astrocyte cultures in vitro. Activation of Piezo1, via the small molecule agonist Yoda1, enhanced Ca
2+ influx in both control and LPS-stimulated astrocytes. Moreover, Yoda1 augmented intracellular Ca2+ oscillations but decreased subsequent Ca2+ influx in response to adenosine triphosphate (ATP) stimulation. Neither blocking nor activating Piezo1 affected cell viability. However, LPS-stimulated astrocyte cultures exposed to the Piezo1 activator, Yoda1, migrated significantly slower than reactive astrocytes treated with the mechanosensitive channel-blocking peptide, GsMTx4. Furthermore, our data show that activating Piezo1 channels inhibits the release of cytokines and chemokines, such as IL-1β, TNFα, and fractalkine (CX3 CL1), from LPS-stimulated astrocyte cultures. Taken together, our results suggest that astrocytic Piezo1 upregulation may act to dampen neuroinflammation and could be a useful drug target for neuroinflammatory disorders of the brain., (© 2019 Wiley Periodicals, Inc.)- Published
- 2020
- Full Text
- View/download PDF
17. Inhibition of the Interaction Between Group I Metabotropic Glutamate Receptors and PDZ-Domain Proteins Prevents Hippocampal Long-Term Depression, but Not Long-Term Potentiation.
- Author
-
Neyman S, Braunewell KH, O'Connell KE, Dev KK, and Manahan-Vaughan D
- Abstract
The group I metabotropic glutamate (mGlu) receptor subtypes, mGlu1 and mGlu5, strongly regulate hippocampal synaptic plasticity. Both harbor PSD-95/discs-large/ZO-1 (PDZ) motifs at their extreme carboxyl terminals, which allow interaction with the PDZ domain of Tamalin, regulate the cell surface expression of group I mGlu receptors, and may modulate their coupling to signaling proteins. We investigated the functional role of this interaction in hippocampal long-term depression (LTD). Acute intracerebral treatment of adult rats with a cell-permeable PDZ-blocking peptide (pep-mGluR-STL), designed to competitively inhibit the interaction between Tamalin and group 1 mGlu receptors, prevented expression of LTD in the hippocampal CA1 region without affecting long-term potentiation (LTP) or basal synaptic transmission. Pep-mGluR-STL prevented facilitation by the group I mGlu receptor agonist, (S)-3,5-Dihydroxyphenylglycine (DHPG), and the mGlu5 agonist, (R,S)-2-chloro-5-Hydroxyphenylglycine (CHPG), of short-term depression (STD) into LTD, suggesting that Tamalin preferentially acts by mediating signaling through mGlu5. These data support that Tamalin is essential for the persistent expression of LTD and that it subserves the effective signaling of group 1 mGlu receptors.
- Published
- 2019
- Full Text
- View/download PDF
18. Infection Augments Expression of Mechanosensing Piezo1 Channels in Amyloid Plaque-Reactive Astrocytes.
- Author
-
Velasco-Estevez M, Mampay M, Boutin H, Chaney A, Warn P, Sharp A, Burgess E, Moeendarbary E, Dev KK, and Sheridan GK
- Abstract
A defining pathophysiological hallmark of Alzheimer's disease (AD) is the amyloid plaque; an extracellular deposit of aggregated fibrillar Aβ
1-42 peptides. Amyloid plaques are hard, brittle structures scattered throughout the hippocampus and cerebral cortex and are thought to cause hyperphosphorylation of tau, neurofibrillary tangles, and progressive neurodegeneration. Reactive astrocytes and microglia envelop the exterior of amyloid plaques and infiltrate their inner core. Glia are highly mechanosensitive cells and can almost certainly sense the mismatch between the normally soft mechanical environment of the brain and very stiff amyloid plaques via mechanosensing ion channels. Piezo1, a non-selective cation channel, can translate extracellular mechanical forces to intracellular molecular signaling cascades through a process known as mechanotransduction . Here, we utilized an aging transgenic rat model of AD (TgF344-AD) to study expression of mechanosensing Piezo1 ion channels in amyloid plaque-reactive astrocytes. We found that Piezo1 is upregulated with age in the hippocampus and cortex of 18-month old wild-type rats. However, more striking increases in Piezo1 were measured in the hippocampus of TgF344-AD rats compared to age-matched wild-type controls. Interestingly, repeated urinary tract infections with Escherichia coli bacteria, a common comorbidity in elderly people with dementia, caused further elevations in Piezo1 channel expression in the hippocampus and cortex of TgF344-AD rats. Taken together, we report that aging and peripheral infection augment amyloid plaque-induced upregulation of mechanoresponsive ion channels, such as Piezo1, in astrocytes. Further research is required to investigate the role of astrocytic Piezo1 in the Alzheimer's brain, whether modulating channel opening will protect or exacerbate the disease state, and most importantly, if Piezo1 could prove to be a novel drug target for age-related dementia.- Published
- 2018
- Full Text
- View/download PDF
19. EBI2 regulates pro-inflammatory signalling and cytokine release in astrocytes.
- Author
-
Rutkowska A, Shimshek DR, Sailer AW, and Dev KK
- Subjects
- Animals, Astrocytes drug effects, Cells, Cultured, Cholesterol analogs & derivatives, Cholesterol pharmacology, Cytokines pharmacology, Dose-Response Relationship, Drug, Humans, Lipopolysaccharides pharmacology, Mice, Mice, Inbred C57BL, Mice, Knockout, NF-kappa B metabolism, Protein Transport drug effects, Protein Transport genetics, Rats, Receptors, G-Protein-Coupled genetics, Signal Transduction drug effects, Astrocytes metabolism, Cytokines metabolism, Receptors, G-Protein-Coupled metabolism, Signal Transduction physiology
- Abstract
The endogenous oxysterol 7α, 25-dihydroxycholesterol (7α25HC) ligand activates the G protein-coupled receptor EBI2 to regulate T cell-dependant antibody response and B cell migration. We have demonstrated that EBI2 is expressed in human and mouse astrocytes, that 7α25HC induces intracellular signalling and astrocyte migration, and that EBI2 plays a role in the crosstalk between astrocytes and macrophages. Recently, we demonstrate that EBI2 regulates myelin development and inhibits LPC-induced demyelination. Here, we show that 7α25HC inhibits LPS- and IL17/TNF-induced pro-inflammatory cytokine release in astrocytes. We observe the following: 1. Human astrocytes treated with IL17/TNF increases the nuclear translocation of NFκB, which is attenuated by pre-treatment with 7α25HC; 2. IL17/TNF increases cell impedance in human astrocytes, which is also attenuated by pre-treatment with 7α25HC; 3. The EBI2 antagonist NIBR189 inhibits these effects of 7α25HC, supporting the role of EBI2; 4. in vivo data corroborate these in vitro findings, showing that EBI2 knock-out (KO) animals display enhanced pro-inflammatory cytokine in response to LPS challenge, in the brain. These results demonstrate a role for oxysterol/EBI2 signalling in attenuating the response of astrocytes to pro-inflammatory signals as well as limiting the levels of pro-inflammatory cytokines in the brain., (Copyright © 2018 The Authors. Published by Elsevier Ltd.. All rights reserved.)
- Published
- 2018
- Full Text
- View/download PDF
20. A novel modelling mechanism of PAEL receptor and GABARAPL2 interaction involved in Parkinson's disease.
- Author
-
Dutta P, Dargahi L, O'Connell KE, Bolia A, Ozkan B, Sailer AW, and Dev KK
- Subjects
- Amino Acid Sequence, Autophagy, Carrier Proteins, Computer Simulation, HEK293 Cells, Humans, Models, Molecular, Protein Interaction Domains and Motifs, Protein Structure, Tertiary, Ubiquitination, Autophagy-Related Protein 8 Family metabolism, Parkinson Disease metabolism, Receptors, G-Protein-Coupled metabolism
- Abstract
Parkin associated endothelin like receptor (PAELR) is G-protein coupled and ubiquitinated by parkin, promoting its degradation. In autosomal recessive Parkinson's disease, mutations in parkin lead to PAELR aggregation in the endoplasmic reticulum (ER), ER stress, neurotoxicity and cell death. We have identified previously that the protein kinase C interacting protein (PICK1) interacts with and regulates the expression and cell toxicity of PAELR. Here, we experimentally identify and provide in-silico modelling of a novel interaction between PAELR and GABARAPL2 (γ-aminobutyrate type A receptor associated protein like 2), which is an autophagosome-specific Ub-like protein implicated in vesicle trafficking and autophagy. We show that the family of GABARAPs interact with the carboxy terminal (ct) of PAELR and find the cysteine rich region (-CCCCCC-EEC) of ct-PAELR interacts with the GABA
A binding site of GABARAPL2. This interaction is modelled by in-slico analysis and confirmed using affinity chromatography, showing Myc-tagged GABARAPL2 is retained by a GST fusion of the ct-PAELR. We also demonstrate that transient transfection of GABARAPL2 in HEK293 cells reduces PAELR expression. This study supports the idea that protein levels of PAELR are likely regulated by a multitude of proteins including parkin, PICK1 and GABARAPL2 via mechanisms that include ubiquitination, proteasomal degradagtion and autophagy., (Copyright © 2018 Elsevier B.V. All rights reserved.)- Published
- 2018
- Full Text
- View/download PDF
21. Sphingosine 1-phosphate receptors regulate TLR4-induced CXCL5 release from astrocytes and microglia.
- Author
-
O'Sullivan SA, O'Sullivan C, Healy LM, Dev KK, and Sheridan GK
- Subjects
- Animals, Astrocytes drug effects, Female, Fingolimod Hydrochloride administration & dosage, Humans, Inflammation Mediators metabolism, Lipopolysaccharides administration & dosage, Male, Mice, Inbred C57BL, Microglia drug effects, RNA, Messenger metabolism, Rats, Wistar, Receptors, Lysosphingolipid agonists, Signal Transduction, Astrocytes metabolism, Chemokine CXCL5 metabolism, Inflammation metabolism, Microglia metabolism, Receptors, Lysosphingolipid metabolism, Toll-Like Receptor 4 metabolism
- Abstract
Sphingosine 1-phosphate receptors (S1PR) are G protein-coupled and compose a family with five subtypes, S1P1R-S1P5R. The drug Gilenya
® (Novartis, Basel, Switzerland) (Fingolimod; FTY720) targets S1PRs and was the first oral therapy for patients with relapsing-remitting multiple sclerosis (MS). The phosphorylated form of FTY720 (pFTY720) binds S1PRs causing initial agonism, then subsequent receptor internalization and functional antagonism. Internalization of S1P1R attenuates sphingosine 1-phosphate (S1P)-mediated egress of lymphocytes from lymph nodes, limiting aberrant immune function in MS. pFTY720 also exerts direct actions on neurons and glial cells which express S1PRs. In this study, we investigated the regulation of pro-inflammatory chemokine release by S1PRs in enriched astrocytes and microglial cultures. Astrocytes and microglia were stimulated with lipopolysaccharide (LPS) and increases in C-X-C motif chemokine 5 (CXCL5), also known as LIX (lipopolysaccharide-induced CXC chemokine) expression were quantified. Results showed that pFTY720 attenuated LPS-induced CXCL5 (LIX) protein release from astrocytes, as did the S1P1R selective agonist, SEW2871. In addition, pFTY720 blocked messenger ribonucleic acid (mRNA) transcription of the chemokines, (i) CXCL5/LIX, (ii) C-X-C motif chemokine 10 (CXCL10) also known as interferon gamma-induced protein 10 (IP10) and (iii) chemokine (C-C motif) ligand 2 (CCL2) also known as monocyte chemoattractant protein 1 (MCP1). Interestingly, inhibition of sphingosine kinase attenuated LPS-induced increases in mRNA levels of all three chemokines, suggesting that LPS-TLR4 (Toll-like receptor 4) signalling may enhance chemokine expression via S1P-S1PR transactivation. Lastly, these observations were not limited to astrocytes since we also found that pFTY720 attenuated LPS-induced release of CXCL5 from microglia. These data highlight a role for S1PR signalling in regulating the levels of chemokines in glial cells and support the notion that pFTY720 efficacy in multiple sclerosis may involve the direct modulation of astrocytes and microglia., (© 2018 International Society for Neurochemistry.)- Published
- 2018
- Full Text
- View/download PDF
22. EBI2 receptor regulates myelin development and inhibits LPC-induced demyelination.
- Author
-
Rutkowska A, Sailer AW, and Dev KK
- Subjects
- Animals, Demyelinating Diseases chemically induced, Lysophosphatidylcholines toxicity, Mice, Mice, Knockout, Myelin Basic Protein biosynthesis, Organ Culture Techniques, Cerebellum metabolism, Demyelinating Diseases metabolism, Myelin Sheath metabolism, Receptors, G-Protein-Coupled metabolism
- Abstract
Background: The G protein-coupled receptor EBI2 (Epstein-Barr virus-induced gene 2) is activated by 7α, 25-dihydroxycholesterol (7α25HC) and plays a role in T cell-dependant antibody response and B cell migration. Abnormal EBI2 signaling is implicated in a range of autoimmune disorders; however, its role in the CNS remains poorly understood., Methods: Here we characterize the role of EBI2 in myelination under normal and pathophysiological conditions using organotypic cerebellar slice cultures and EBI2 knock-out (KO) animals., Results: We find that MBP expression in brains taken from EBI2 KO mice is delayed compared to those taken from wild type (WT) mice. In agreement with these in vivo findings, we show that antagonism of EBI2 reduces MBP expression in vitro. Importantly, we demonstrate that EBI2 activation attenuates lysolecithin (LPC)-induced demyelination in mouse organotypic slice cultures. Moreover, EBI2 activation also inhibits LPC-mediated release of pro-inflammatory cytokines such as IL6 and IL1β in cerebellar slices., Conclusions: These results, for the first time, display a role for EBI2 in myelin development and protection from demyelination under pathophysiological conditions and suggest that modulation of this receptor may be beneficial in neuroinflammatory and demyelinating disorders such as multiple sclerosis.
- Published
- 2017
- Full Text
- View/download PDF
23. Phospholipase A2 is involved in galactosylsphingosine-induced astrocyte toxicity, neuronal damage and demyelination.
- Author
-
Misslin C, Velasco-Estevez M, Albert M, O'Sullivan SA, and Dev KK
- Subjects
- Animals, Cells, Cultured, Enzyme Inhibitors pharmacology, Humans, Mice, Phospholipases A2 drug effects, Astrocytes physiology, Demyelinating Diseases, Neurons pathology, Phospholipases A2 physiology, Psychosine physiology
- Abstract
Krabbe disease is a fatal rare inherited lipid storage disorder affecting 1:100,000 births. This illness is caused by mutations in the galc gene encoding for the enzyme galactosylceramidase (GALC). Dysfunction of GALC has been linked to the toxic build-up of the galactolipid, galactosylsphingosine (psychosine), which induces cell death of oligodendrocytes. Previous studies show that phospholipase A2 (PLA2) may play a role in psychosine induce cell death. Here, we demonstrate that non-selective inhibition of cPLA2/sPLA2 and selective inhibition of cPLA2, but not sPLA2, also attenuates psychosine-induced cell death of human astrocytes. This study shows that extracellular calcium is required for psychosine induced cell death, but intracellular calcium release, reactive oxygen species or release of soluble factors are not involved. These findings suggest a cell autonomous effect, at least in human astrocytes. Supporting a role for PLA2 in psychosine-induced cell death of oligodendrocytes and astrocytes, the results show inhibition of PLA2 attenuates psychosine-induced decrease in the expression of astrocyte marker vimentin as well as myelin basic protein (MBP), myelin oligodendrocyte glycoprotein (MOG) and the neuronal marker SMI-32 in organotypic slice cultures. These findings provide further mechanistic details of psychosine-induced death of glia and suggest a role for PLA2 in the process. This work also supports the proposal that novel drugs for Krabbe disease may require testing on astrocytes as well as oligodendrocytes for more holistic prediction of pre-clinical and clinical efficacy.
- Published
- 2017
- Full Text
- View/download PDF
24. The chemokine fractalkine (CX3CL1) attenuates H 2 O 2 -induced demyelination in cerebellar slices.
- Author
-
O'Sullivan SA and Dev KK
- Subjects
- Animals, Animals, Newborn, CX3C Chemokine Receptor 1 metabolism, Catalase toxicity, Cell Death drug effects, Female, Gliosis chemically induced, Gliosis prevention & control, Glucose Oxidase toxicity, Male, Mice, Mice, Inbred C57BL, Myelin Basic Protein metabolism, Myelin-Oligodendrocyte Glycoprotein metabolism, Neurofilament Proteins metabolism, Organ Culture Techniques, Reactive Oxygen Species metabolism, Cerebellum drug effects, Cerebellum pathology, Chemokine CX3CL1 therapeutic use, Demyelinating Diseases chemically induced, Demyelinating Diseases prevention & control, Hydrogen Peroxide toxicity
- Abstract
Background: Fractalkine/CX3CR1 signalling has been implicated in many neurodegenerative and neurological diseases of the central nervous system (CNS). This signalling pathway plays an important role in regulating reactive oxygen species (ROS), as well as itself being altered in conditions of oxidative stress. Here, we investigated the effects of recombinant fractalkine (rCX3CL1) in models of hydrogen peroxide (H
2 O2 )-induced demyelination and astrocyte toxicity, within organotypic cerebellar slice cultures., Methods: Organotypic cerebellar slice cultures were generated from postnatal day 10 C57BL/6J mice to assess myelination. Immunohistochemistry was used to measure the degree of myelination. Fluorescent images were obtained using a leica SP8 confocal microscope and data analysed using ImageJ software., Results: We show here, for the first time, that rCX3CL1 significantly attenuated bolus H2 O2 -induced demyelination as measured by expression of myelin basic protein (MBP) and attenuated reduced vimentin expression. Using the GOX-CAT system to continuously generate low levels of H2 O2 and induce demyelination, we observed similar protective effects of rCX3CL1 on MBP and MOG fluorescence, although in this model, the decrease in vimentin expression was not altered., Conclusions: This data indicates possible protective effects of fractalkine signalling in oxidative stress-induced demyelination in the central nervous system. This opens up the possibility of fractalkine receptor (CX3CR1) modulation as a potential new target for protecting against oxidative stress-induced demyelination in both inflammatory and non-inflammatory nervous system disorders.- Published
- 2017
- Full Text
- View/download PDF
25. 6th Annual Meeting Frontiers in Neurology 2016.
- Author
-
Dev KK
- Published
- 2017
- Full Text
- View/download PDF
26. Demyelination induced by oxidative stress is regulated by sphingosine 1-phosphate receptors.
- Author
-
O'Sullivan SA, Velasco-Estevez M, and Dev KK
- Subjects
- Animals, Animals, Newborn, Astrocytes drug effects, Astrocytes physiology, Brain cytology, Calcium-Binding Proteins, Cells, Cultured, Cerebellum drug effects, DNA-Binding Proteins metabolism, Demyelinating Diseases drug therapy, Female, Fetus, Fingolimod Hydrochloride therapeutic use, Gene Expression Regulation drug effects, Humans, Hydrogen Peroxide toxicity, Immunosuppressive Agents therapeutic use, Male, Mice, Mice, Inbred C57BL, Microfilament Proteins, Myelin-Oligodendrocyte Glycoprotein metabolism, Neurofilament Proteins metabolism, Oxidative Stress drug effects, Receptors, Lysosphingolipid genetics, Repressor Proteins metabolism, Zebrafish Proteins metabolism, Demyelinating Diseases etiology, Demyelinating Diseases metabolism, Oxidative Stress physiology, Receptors, Lysosphingolipid metabolism
- Abstract
Oxidative stress is a pathological condition defined as an imbalance between production and removal of reactive oxygen species. This process causes structural cell damage, disrupts DNA repair and induces mitochondrial dysfunction. Many in vitro studies have used direct bolus application of H
2 O2 to investigate the role of oxidative stress in cell culture. In this study, using mouse organotypic cerebellar slice cultures, the effects of H2 O2 -induced oxidative stress on myelination state were examined, using bolus concentrations of H2 O2 (0.1-1 mM) and low-continuous H2 O2 (∼20 μM) generated from glucose oxidase and catalase (GOX-CAT). Using these models, the potential therapeutic effects of pFTY720, an oral therapy used in multiple sclerosis, was also examined. We found bolus treatment of H2 O2 (0.5 mM) and, for the first time, low-continuous H2 O2 (GOX-CAT) to induce demyelination in organotypic slices. Both bolus H2 O2 and GOX-CAT treatments significantly decreased vimentin expression in these slice cultures as well as increased cell death in isolated astrocyte cultures. Importantly, pre-treatment with pFTY720 significantly attenuated both bolus H2 O2 and GOX-CAT-induced demyelination and the GOX-CAT-induced decrease in vimentin in cerebellar slices, without altering levels of the proinflammatory cytokines such as IL-6 and CX3CL1. We also observed increased SMI-32 immunoreactivity in the white matter tract induced by GOX-CAT indicating axonal damage, which was remarkably attenuated by pFTY720. Taken together, this data establishes a novel GOX-CAT model of demyelination and demonstrates that pFTY720 can act independently of inflammatory cytokines to attenuate decreases in vimentin, as well as axonal damage and demyelination induced by oxidative stress., (© 2017 Wiley Periodicals, Inc.)- Published
- 2017
- Full Text
- View/download PDF
27. Sphingosine-1-phosphate receptor therapies: Advances in clinical trials for CNS-related diseases.
- Author
-
O'Sullivan S and Dev KK
- Subjects
- Animals, Clinical Trials as Topic, Humans, Lysophospholipids metabolism, Sphingosine analogs & derivatives, Sphingosine metabolism, Central Nervous System Agents pharmacology, Central Nervous System Agents therapeutic use, Central Nervous System Diseases drug therapy, Central Nervous System Diseases metabolism, Receptors, Lysosphingolipid metabolism
- Abstract
The family of sphingosine-1-phosphate receptors (S1PRs) are G protein-coupled and comprise of five subtypes, S1P
1 -S1P5 . These receptors are activated by the sphingolipid ligand, S1P, which is produced from the phosphorylation of sphingosine by sphingosine kinases. The activation of S1PRs modulates a host of cellular processes such as cell proliferation, migration and survival. These receptors are targeted by the drug fingolimod, a first in class oral therapy for multiple sclerosis. Importantly, S1PRs have also been implicated, in cellular experiments, pre-clinical studies and clinical trials in a range of other neurodegenerative diseases, neurological disorders and psychiatric illnesses, where S1PR drugs are proving beneficial. Overall, studies now highlight the importance of S1PRs as targets for modulating a variety of debilitating brain-related diseases. Here, we review the role of S1PRs in these illnesses. This article is part of the Special Issue entitled 'Lipid Sensing G Protein-Coupled Receptors in the CNS'., (Copyright © 2016 Elsevier Ltd. All rights reserved.)- Published
- 2017
- Full Text
- View/download PDF
28. Lipid sensing G protein-coupled receptors in the CNS.
- Author
-
Dev KK and Irving AJ
- Subjects
- Animals, Humans, Central Nervous System metabolism, Lipids, Receptors, G-Protein-Coupled physiology
- Published
- 2017
- Full Text
- View/download PDF
29. In vitro and ex vivo models of multiple sclerosis.
- Author
-
Madill M, Fitzgerald D, O'Connell KE, Dev KK, Shen S, and FitzGerald U
- Subjects
- Animals, Brain, Cells, Cultured, Drug Evaluation, Preclinical methods, Humans, Oligodendroglia, Tissue Culture Techniques, Models, Biological, Multiple Sclerosis
- Abstract
Multiple sclerosis (MS) is an autoimmune disorder of the central nervous system (CNS). Current therapies suppress a misdirected myelin-destructive immune response. To combat the progressive, neurodestructive phase of MS, the therapeutic research focus is currently on compounds that might boost the endogenous potential of the brain to remyelinate axons, thereby achieving lesion repair. Here, we describe the testing of fingolimod on cultures of oligodendrocytes (OLs) and organotypic brain slices. We detail the protocols, pros, and cons of these in vitro and ex vivo approaches, along with the potential benefit of exploiting skin-punch biopsies from patients with MS, before concluding with a summary of future developments., (Copyright © 2016 Elsevier Ltd. All rights reserved.)
- Published
- 2016
- Full Text
- View/download PDF
30. Fractalkine shedding is mediated by p38 and the ADAM10 protease under pro-inflammatory conditions in human astrocytes.
- Author
-
O'Sullivan SA, Gasparini F, Mir AK, and Dev KK
- Subjects
- Cells, Cultured, Chemokine CX3CL1 genetics, Culture Media, Serum-Free pharmacology, Dose-Response Relationship, Drug, Enzyme Inhibitors pharmacology, Humans, Matrix Metalloproteinases metabolism, RNA, Messenger metabolism, Time Factors, ADAM10 Protein metabolism, Amyloid Precursor Protein Secretases metabolism, Astrocytes drug effects, Astrocytes metabolism, Chemokine CX3CL1 metabolism, Cytokines pharmacology, Membrane Proteins metabolism, p38 Mitogen-Activated Protein Kinases metabolism
- Abstract
Background: The fractalkine (CX3CR1) ligand is expressed in astrocytes and reported to be neuroprotective. When cleaved from the membrane, soluble fractalkine (sCX3CL1) activates the receptor CX3CR1. Although somewhat controversial, CX3CR1 is reported to be expressed in neurons and microglia. The membrane-bound form of CX3CL1 additionally acts as an adhesion molecule for microglia and infiltrating white blood cells. Much research has been done on the role of fractalkine in neuronal cells; however, little is known about the regulation of the CX3CL1 ligand in astrocytes., Methods: The mechanisms involved in the up-regulation and cleavage of CX3CL1 from human astrocytes were investigated using immunocytochemistry, Q-PCR and ELISA. All statistical analysis was performed using GraphPad Prism 5., Results: A combination of ADAM17 (TACE) and ADAM10 protease inhibitors was found to attenuate IL-1β-, TNF-α- and IFN-γ-induced sCX3CL1 levels in astrocytes. A specific ADAM10 (but not ADAM17) inhibitor also attenuated these effects, suggesting ADAM10 proteases induce release of sCX3CL1 from stimulated human astrocytes. A p38 MAPK inhibitor also attenuated the levels of sCX3CL1 upon treatment with IL-1β, TNF-α or IFN-γ. In addition, an IKKβ inhibitor significantly reduced the levels of sCX3CL1 induced by IL-1β or TNF-α in a concentration-dependent manner, suggesting a role for the NF-kB pathway., Conclusions: In conclusion, this study shows that the release of soluble astrocytic fractalkine is regulated by ADAM10 proteases with p38 MAPK also playing a role in the fractalkine shedding event. These findings are important for understanding the role of CX3CL1 in healthy and stimulated astrocytes and may benefit our understanding of this pathway in neuro-inflammatory and neurodegenerative diseases.
- Published
- 2016
- Full Text
- View/download PDF
31. The EBI2 signalling pathway plays a role in cellular crosstalk between astrocytes and macrophages.
- Author
-
Rutkowska A, O'Sullivan SA, Christen I, Zhang J, Sailer AW, and Dev KK
- Subjects
- Animals, Astrocytes immunology, Cell Line, Cell Movement, Chromatography, High Pressure Liquid, Gene Expression Profiling, Gene Expression Regulation, Lipopolysaccharides immunology, Macrophages immunology, Mice, Oxysterols metabolism, Proteomics methods, Tandem Mass Spectrometry, Transcriptome, Astrocytes metabolism, Cell Communication, Macrophages metabolism, Receptors, G-Protein-Coupled metabolism, Signal Transduction
- Abstract
EBI2 is a G protein-coupled receptor activated by oxysterol 7α, 25-dihydroxycholesterol (7α25HC) and regulates T cell-dependant antibody response and B cell migration. We recently found EBI2 is expressed in human astrocytes, regulates intracellular signalling and modulates astrocyte migration. Here, we report that LPS treatment of mouse astrocytes alters mRNA levels of EBI2 and oxysterols suggesting that the EBI2 signalling pathway is sensitive to LPS-mediated immune challenge. We also find that conditioned media obtained from LPS-stimulated mouse astrocytes induces macrophage migration, which is inhibited by the EBI2 antagonist NIBR189. These results demonstrate a role for the EBI2 signalling pathway in astrocytes as a sensor for immune challenge and for communication with innate immune cells such as macrophages.
- Published
- 2016
- Full Text
- View/download PDF
32. "5th Annual Meeting Frontiers in Neurology 2015".
- Author
-
Dev KK
- Published
- 2016
- Full Text
- View/download PDF
33. The dual S1PR1/S1PR5 drug BAF312 (Siponimod) attenuates demyelination in organotypic slice cultures.
- Author
-
O'Sullivan C, Schubart A, Mir AK, and Dev KK
- Subjects
- Animals, Animals, Newborn, Calcium Signaling drug effects, Calcium Signaling genetics, Humans, Immunosuppressive Agents pharmacology, In Vitro Techniques, Indans pharmacology, Interleukin-6 metabolism, Lysophosphatidylcholines pharmacology, Mice, Myelin Basic Protein metabolism, Organ Culture Techniques, Oxadiazoles pharmacology, Protein Transport drug effects, Receptors, Lysosphingolipid agonists, Receptors, Lysosphingolipid antagonists & inhibitors, Receptors, Lysosphingolipid metabolism, Thiophenes pharmacology, Time Factors, beta-Alanine analogs & derivatives, beta-Alanine pharmacology, eIF-2 Kinase metabolism, Anti-Inflammatory Agents pharmacology, Astrocytes drug effects, Azetidines pharmacology, Benzyl Compounds pharmacology, Cerebellum cytology, Demyelinating Diseases drug therapy
- Abstract
Background: BAF312 (Siponimod) is a dual agonist at the sphingosine-1 phosphate receptors, S1PR1 and S1PR5. This drug is currently undergoing clinical trials for the treatment of secondary progressive multiple sclerosis (MS). Here, we investigated the effects of BAF312 on isolated astrocyte and microglia cultures as well as in slice culture models of demyelination., Methods: Mouse and human astrocytes were treated with S1PR modulators and changes in the levels of pERK, pAkt, and calcium signalling as well as S1PR1 internalization and cytokine levels was investigated using Western blotting, immunochemistry, ELISA and confocal microscopy. Organotypic slice cultures were prepared from the cerebellum of 10-day-old mice and treated with lysophosphatidylcholine (LPC), psychosine and/or S1PR modulators, and changes in myelination states were measured by fluorescence of myelin basic protein and neurofilament H., Results: BAF312 treatment of human and mouse astrocytes activated pERK, pAKT and Ca(2+) signalling as well as inducing S1PR1 internalization. Notably, activation of S1PR1 increased pERK and pAKT in mouse astrocytes while both S1PR1 and S1PR3 equally increased pERK and pAKT in human astrocytes, suggesting that the coupling of S1PR1 and S1PR3 to pERK and pAKT differ in mouse and human astrocytes. We also observed that BAF312 moderately attenuated lipopolysaccharide (LPS)- or TNFα/IL17-induced levels of IL6 in both astrocyte and microglia cell cultures. In organotypic slice cultures, BAF312 reduced LPC-induced levels of IL6 and attenuated LPC-mediated demyelination. We have shown previously that the toxic lipid metabolite psychosine induces demyelination in organotypic slice cultures, without altering the levels of cytokines, such as IL6. Importantly, psychosine-induced demyelination was also attenuated by BAF312., Conclusions: Overall, this study suggests that BAF312 can modulate glial cell function and attenuate demyelination, highlighting this drug as a further potential therapy in demyelinating disorders, beyond MS.
- Published
- 2016
- Full Text
- View/download PDF
34. The Role of the Oxysterol/EBI2 Pathway in the Immune and Central Nervous Systems.
- Author
-
Rutkowska A, Dev KK, and Sailer AW
- Subjects
- Adaptive Immunity, Animals, Astrocytes metabolism, Cytochrome P450 Family 7 metabolism, Humans, Inflammation metabolism, Neurodegenerative Diseases metabolism, Receptors, G-Protein-Coupled genetics, Signal Transduction, Steroid Hydroxylases metabolism, Central Nervous System metabolism, Immune System metabolism, Oxysterols metabolism, Receptors, G-Protein-Coupled metabolism
- Abstract
Oxysterols are pleiotropic messengers interacting with multiple receptor systems. One of the cognate receptors for oxysterols is EBI2, a G protein-coupled receptor highly expressed in the cells of the immune system. Here we discuss the receptor's role in the adapted immunity and inflammation as well as the receptor's expression and function in the CNS with the focus on astrocytes. We also discuss expression and signalling of oxysterol-producing enzymes such as CH25H and CYP7B1 in the CNS and the immune system. These steps will help to elucidate a possible role for this pathway in the physiology of the central and peripheral nervous system and its possible link to human disease.
- Published
- 2016
- Full Text
- View/download PDF
35. Editorial (Thematic Issue: Common Receptor Signalling in Glial & Immune Cells).
- Author
-
Dev KK
- Subjects
- Astrocytes physiology, Cell Communication physiology, Immune System cytology, Neuroglia physiology, Receptors, Immunologic physiology
- Published
- 2016
- Full Text
- View/download PDF
36. Galactosylsphingosine (psychosine)-induced demyelination is attenuated by sphingosine 1-phosphate signalling.
- Author
-
O'Sullivan C and Dev KK
- Subjects
- Animals, Apoptosis drug effects, Astrocytes drug effects, Astrocytes metabolism, Demyelinating Diseases drug therapy, Demyelinating Diseases metabolism, Fingolimod Hydrochloride pharmacology, Fingolimod Hydrochloride therapeutic use, Humans, Leukodystrophy, Globoid Cell drug therapy, Leukodystrophy, Globoid Cell metabolism, Lipopolysaccharides pharmacology, Lysophospholipids, Mice, Microscopy, Fluorescence, Psychosine therapeutic use, Rats, Signal Transduction drug effects, Sphingosine analogs & derivatives, Demyelinating Diseases chemically induced, Psychosine pharmacology
- Abstract
Globoid cell leukodystrophy (Krabbe disease) is a rare infantile neurodegenerative disorder. Krabbe disease is caused by deficiency in the lysosomal enzyme galactocerebrosidase (GALC) resulting in accumulation, in the micromolar range, of the toxic metabolite galactosylsphingosine (psychosine) in the brain. Here we find that psychosine induces human astrocyte cell death probably via an apoptotic process in a concentration- and time-dependent manner (EC50 ∼ 15 μM at 4 h). We show these effects of psychosine are attenuated by pre-treatment with the sphingosine 1-phosphate receptor agonist pFTY720 (fingolimod) (IC50 ∼ 100 nM). Psychosine (1 μM, 10 μM) also enhances LPS-induced (EC50 ∼ 100 ng/ml) production of pro-inflammatory cytokines in mouse astrocytes, which is also attenuated by pFTY720 (1 μM). Most notably, for the first time, we show that psychosine, at a concentration found in the brains of patients with Krabbe disease (EC50 ∼ 100 nM), directly induces demyelination in mouse organotypic cerebellar slices in a manner that is independent of pro-inflammatory cytokine response and that pFTY720 (0.1 nM) significantly inhibits. These results support the idea that psychosine is a pathogenic agent in Krabbe disease and suggest that sphingosine 1-phosphate signalling could be a potential drug target for this disorder., (© 2015. Published by The Company of Biologists Ltd.)
- Published
- 2015
- Full Text
- View/download PDF
37. Increased interleukin 23 (IL23) levels in schizophrenia patients treated with depot antipsychotic medication.
- Author
-
O'Connell KE, Thakore J, and Dev KK
- Subjects
- Adult, Delayed-Action Preparations, Female, Humans, Male, Middle Aged, Time Factors, Young Adult, Antipsychotic Agents therapeutic use, Interleukin-23 Subunit p19 blood, Schizophrenia blood, Schizophrenia drug therapy
- Abstract
Background: Schizophrenia is a mental disorder, where genetic and environmental factors contribute to disease onset and progression. The immune system appears to play a role in schizophrenia, where altered cytokines levels and autoantibodies have been described. Notably however, to our knowledge, IL23 levels have not before been measured in schizophrenia patients treated with depot medication., Methods: We examined IL23 levels in serum samples obtained from patients with schizophrenia, treated with depot medication (n=35) compared with healthy controls (n=38) and correlated these levels with treatment time, patient age and illness severity., Results: IL23 levels were raised in depot treated groups compared with healthy controls. No correlation was observed, however, between IL23 levels and treatment time, patient age or illness severity., Conclusions: IL23 levels are raised in schizophrenia patients prescribed with depot medication, supporting the role of aberrant cytokine signalling in schizophrenia., (Copyright © 2015. Published by Elsevier Ltd.)
- Published
- 2015
- Full Text
- View/download PDF
38. EBI2 regulates intracellular signaling and migration in human astrocyte.
- Author
-
Rutkowska A, Preuss I, Gessier F, Sailer AW, and Dev KK
- Subjects
- Analysis of Variance, Animals, Animals, Newborn, Area Under Curve, Calcium Signaling physiology, Cell Differentiation, Cell Movement genetics, Cerebral Cortex cytology, Cholesterol pharmacokinetics, Extracellular Signal-Regulated MAP Kinases metabolism, Flow Cytometry, Glial Fibrillary Acidic Protein metabolism, Humans, Mice, Mice, Inbred C57BL, Mice, Knockout, Receptors, G-Protein-Coupled antagonists & inhibitors, Receptors, G-Protein-Coupled genetics, Signal Transduction, Transfection, Astrocytes metabolism, Cell Movement physiology, Receptors, G-Protein-Coupled metabolism
- Abstract
The G protein-coupled receptor EBI2 (Epstein-Barr virus-induced gene 2) is activated by 7α, 25-dihydroxycholesterol (7α25HC) and plays a role in T cell-dependant antibody response and B cell migration. Aberrant EBI2 signaling is implicated in a range of autoimmune disorders however its role in the CNS remains unknown. Here we characterize the functional role of EBI2 in GLIA cells using primary human astrocytes and EBI2 knockout animals. We find human and mouse astrocytes express EBI2 and the enzymes necessary for synthesis and degradation of 7α25HC. In astrocytes, EBI2 activation stimulates ERK phosphorylation, Ca(2+) signaling and induces cellular migration. These results, for the first time, demonstrate a role for EBI2 in astrocyte function and suggest that modulation of this receptor may be beneficial in neuroinflammatory disorders., (© 2014 Wiley Periodicals, Inc.)
- Published
- 2015
- Full Text
- View/download PDF
39. The use of cytokine signature patterns: separating drug naïve, interferon and natalizumab-treated multiple sclerosis patients.
- Author
-
O'Connell KE, Mok T, Sweeney B, Ryan AM, and Dev KK
- Subjects
- Adult, Biomarkers blood, Female, Humans, Immunoblotting, Male, Multiple Sclerosis, Relapsing-Remitting blood, Natalizumab, Statistics, Nonparametric, Antibodies, Monoclonal, Humanized administration & dosage, Cytokines blood, Interferon-beta administration & dosage, Multiple Sclerosis, Relapsing-Remitting drug therapy, Multiple Sclerosis, Relapsing-Remitting immunology
- Abstract
Multiple sclerosis (MS) is an inflammatory illness characterized by demyelination and axonal neurodegeneration. Here, we used serum samples from MS patients to demonstrate if "cytokine signature" patterns can separate different patient groups better than using single cytokines. In this case, we used cytokine profiling to demonstrate if "cytokine signature" patterns can separate MS patients treated with interferon or natalizumab from drug naïve patients. Serum levels of eight individual cytokines (TNFα, IFNγ, S100B, IL-1β, IL-6, IL-8, IL-17 and IL-23) in MS patients treated with interferons (n = 11) and natalizumab (n = 14) were measured and, in general, showed reduced levels compared to drug naïve MS patients (n = 12). More evident changes were seen when analyzing "cytokine signatures" (i.e. summed value of all eight cytokines), which showed that patients treated with natalizumab and interferons showed significantly reduced cytokine signature levels than drug naïve MS patients. Moreover, patients treated with natalizumab were separated from drug naïve patients by almost 100% fidelity and that patients treated with natalizumab also had reduced levels of pro-inflammatory cytokines compared to patients treated with interferon. Overall, this study provides an example showing that the use of "cytokine signatures" may provide benefits over the analysis of single cytokines for the development of potential biomarkers.
- Published
- 2014
- Full Text
- View/download PDF
40. The protein interacting with C-kinase (PICK1) interacts with and attenuates parkin-associated endothelial-like (PAEL) receptor-mediated cell death.
- Author
-
Dutta P, O'Connell KE, Ozkan SB, Sailer AW, and Dev KK
- Subjects
- Amino Acid Sequence, Animals, Blotting, Western, Carrier Proteins metabolism, Cells, Cultured, Cytoskeletal Proteins, DNA, Complementary biosynthesis, DNA, Complementary genetics, Female, Glutathione Transferase metabolism, HEK293 Cells, Humans, Immunohistochemistry, Immunoprecipitation, Models, Molecular, Molecular Sequence Data, Nerve Tissue Proteins metabolism, Nuclear Proteins metabolism, PDZ Domains, Polymerase Chain Reaction, Protein Conformation, Rats, Rats, Wistar, Receptors, G-Protein-Coupled metabolism, Carrier Proteins physiology, Cell Death physiology, Nerve Tissue Proteins physiology, Nuclear Proteins physiology, Receptors, G-Protein-Coupled physiology
- Abstract
The parkin-associated endothelial-like receptor (PAELR, GPR37) is an orphan G protein-coupled receptor that interacts with and is degraded by parkin-mediated ubiquitination. Mutations in parkin are thought to result in PAELR accumulation and increase neuronal cell death in Parkinson's disease. In this study, we find that the protein interacting with C-kinase (PICK1) interacts with PAELR. Specifically, the Postsynaptic density protein-95/Discs large/ZO-1 (PDZ) domain of PICK1 interacted with the last three residues of the c-terminal (ct) located PDZ motif of PAELR. Pull-down assays indicated that recombinant and native PICK1, obtained from heterologous cells and rat brain tissue, respectively, were retained by a glutathione S-transferase fusion of ct-PAELR. Furthermore, coimmunoprecipitation studies isolated a PAELR-PICK1 complex from transiently transfected cells. PICK1 interacts with parkin and our data showed that PICK1 reduces PAELR expression levels in transiently transfected heterologous cells compared to a PICK1 mutant that does not interact with PAELR. Finally, PICK1 over-expression in HEK293 cells reduced cell death induced by PAEALR over-expression during rotenone treatment and these effects of PICK1 were attenuated during inhibition of the proteasome. These results suggest a role for PICK1 in preventing PAELR-induced cell toxicity., (© 2014 International Society for Neurochemistry.)
- Published
- 2014
- Full Text
- View/download PDF
41. The premium of a big pharma license deal.
- Author
-
O'Connell KE, Frei P, and Dev KK
- Subjects
- Internationality, Drug Industry economics, Licensure economics, Pharmaceutical Preparations economics, Technology Transfer
- Published
- 2014
- Full Text
- View/download PDF
42. Fingolimod attenuates splenocyte-induced demyelination in cerebellar slice cultures.
- Author
-
Pritchard AJ, Mir AK, and Dev KK
- Subjects
- Animals, Coculture Techniques, Cytokines metabolism, Disease Models, Animal, Female, Fingolimod Hydrochloride, Inflammation Mediators metabolism, Mice, Mice, Transgenic, Myelin-Oligodendrocyte Glycoprotein immunology, Phenotype, Sphingosine pharmacology, T-Lymphocytes immunology, T-Lymphocytes metabolism, Tissue Culture Techniques, Cerebellum immunology, Cerebellum pathology, Demyelinating Diseases immunology, Immunosuppressive Agents pharmacology, Propylene Glycols pharmacology, Sphingosine analogs & derivatives, Spleen cytology, Spleen immunology
- Abstract
The family of sphingosine-1-phosphate receptors (S1PRs) is G-protein-coupled, comprised of subtypes S1PR1-S1PR5 and activated by the endogenous ligand S1P. The phosphorylated version of Fingolimod (pFTY720), an oral therapy for multiple sclerosis (MS), induces S1PR1 internalisation in T cells, subsequent insensitivity to S1P gradients and sequestering of these cells within lymphoid organs, thus limiting immune response. S1PRs are also expressed in neuronal and glial cells where pFTY720 is suggested to directly protect against lysolecithin-induced deficits in myelination state in organotypic cerebellar slices. Of note, the effect of pFTY720 on immune cells already migrated into the CNS, prior to treatment, has not been well established. We have previously found that organotypic slice cultures do contain immune cells, which, in principle, could also be regulated by pFTY720 to maintain levels of myelin. Here, a mouse organotypic cerebellar slice and splenocyte co-culture model was thus used to investigate the effects of pFTY720 on splenocyte-induced demyelination. Spleen cells isolated from myelin oligodendrocyte glycoprotein immunised mice (MOG-splenocytes) or from 2D2 transgenic mice (2D2-splenocytes) both induced demyelination when co-cultured with mouse organotypic cerebellar slices, to a similar extent as lysolecithin. As expected, in vivo treatment of MOG-immunised mice with FTY720 inhibited demyelination induced by MOG-splenocytes. Importantly, in vitro treatment of MOG- and 2D2-splenocytes with pFTY720 also attenuated demyelination caused by these cells. In addition, while in vitro treatment of 2D2-splenocytes with pFTY720 did not alter cell phenotype, pFTY720 inhibited the release of the pro-inflammatory cytokines such as interferon gamma (IFNγ) and interleukin 6 (IL6) from these cells. This work suggests that treatment of splenocytes by pFTY720 attenuates demyelination and reduces pro-inflammatory cytokine release, which likely contributes to enhanced myelination state induced by pFTY720 in organotypic cerebellar slices.
- Published
- 2014
- Full Text
- View/download PDF
43. Pro-inflammatory cytokine levels are raised in female schizophrenia patients treated with clozapine.
- Author
-
O'Connell KE, Thakore J, and Dev KK
- Subjects
- Adult, Aged, Body Mass Index, Female, Humans, Male, Middle Aged, Psychiatric Status Rating Scales, Statistics as Topic, Statistics, Nonparametric, Young Adult, Antipsychotic Agents therapeutic use, Clozapine therapeutic use, Cytokines blood, Schizophrenia blood, Schizophrenia drug therapy, Sex Characteristics
- Abstract
Background: We have previously shown that the neurotrophic factor, S100B, is raised in serum samples of female patients with schizophrenia, but not male patients, compared to controls, and this may be associated with raised BMI. Here we analysed the levels of additional proinflammatory cytokines in patients with schizophrenia to further investigate these gender differences., Methods: The levels of six cytokines (IL1β, IL6, IL8, IL17, IL23, TNFα) were measured in serum samples obtained from patients with schizophrenia, treated with clozapine (n=91) and compared with healthy controls (n=50). Individual cytokine levels were measured using dot-immunoblotting methods and a 'cytokine signature' was also generated by summing all 6 cytokines. Treatment time, patient age, gender, illness severity and metabolic parameters were also measured., Results: The levels of proinflammatory cytokines and BMI were significantly raised in female, but not male, patients treated with clozapine compared to healthy controls. Compared to individual cytokines, the 'cytokine signature' analysis showed less scatter of data although this 'cytokine signature' method did not improve separation of individual patients and controls., Conclusions: This study supports previous findings that raised BMI, which is likely associated with increased number of adipocytes, may contribute to increased cytokine serum concentrations in females., (Copyright © 2014 Elsevier B.V. All rights reserved.)
- Published
- 2014
- Full Text
- View/download PDF
44. Targeting S1P receptors in experimental autoimmune encephalomyelitis in mice improves early deficits in locomotor activity and increases ultrasonic vocalisations.
- Author
-
Sheridan GK and Dev KK
- Subjects
- Animals, Encephalomyelitis, Autoimmune, Experimental pathology, Encephalomyelitis, Autoimmune, Experimental prevention & control, Female, Fingolimod Hydrochloride, Immunosuppressive Agents pharmacology, Lysophospholipids metabolism, Mice, Mice, Inbred C57BL, Motor Activity drug effects, Propylene Glycols pharmacology, Receptors, Lysosphingolipid genetics, Receptors, Lysosphingolipid metabolism, Sphingosine analogs & derivatives, Sphingosine metabolism, Sphingosine pharmacology, Vocalization, Animal drug effects, Disease Models, Animal, Encephalomyelitis, Autoimmune, Experimental metabolism, Motor Activity physiology, Receptors, Lysosphingolipid antagonists & inhibitors, Vocalization, Animal physiology
- Abstract
Fingolimod (FTY720) is an oral therapy for relapsing remitting multiple sclerosis (MS) and targets sphingosine 1-phosphate receptors (S1PRs). FTY720 also rescues animals from experimental autoimmune encephalomyelitis (EAE), an animal model of MS. The protective effects of FTY720 in EAE are primarily scored manually by examining weight loss and limb paralysis that begins around 10-12 days after immunisation. To our knowledge, pre-clinical effects of FTY720 on animal behaviour early in EAE have not been explored. Here, we developed an automated behaviour monitoring system to examine the early effects of FTY720 on subtle pre-symptomatic behaviour of mice induced with EAE. Our automated home-cage monitoring system (AHC-MS) enabled non-contact detection of movement and ultrasonic vocalisations (USVs) of mice induced with EAE, thus allowing detection of subtle changes in mouse behaviour before paralysis occurs. Mice receiving FTY720 emit longer USVs and display higher levels of motor activity than vehicle-treated EAE mice before clinical symptoms become apparent. Importantly, this study promotes the 3Rs ethics (replacement, reduction and refinement) in the EAE animal model and may also improve pre-screening of potentially novel MS therapies. In addition, this is the first report showing the early effects of FTY720 in EAE which underscores its protective effects.
- Published
- 2014
- Full Text
- View/download PDF
45. The expression of neuronal sorting nexin 8 (SNX8) exacerbates abnormal cholesterol levels.
- Author
-
Muirhead G and Dev KK
- Subjects
- Androstenes pharmacology, Animals, Anticholesteremic Agents pharmacology, Astrocytes drug effects, Astrocytes metabolism, Cells, Cultured, Female, HEK293 Cells, Humans, Lovastatin pharmacology, Neurons drug effects, Organ Specificity, Rats, Rats, Wistar, Sorting Nexins genetics, Sterol Regulatory Element Binding Proteins metabolism, Cholesterol metabolism, Neurons metabolism, Sorting Nexins metabolism
- Abstract
SNX8 is a PX-BAR domain sub-family of sorting nexins (SNXs), which is reported as a β-amyloid (Aβ) toxicity enhancer and associated with Alzheimer's disease. We have also described SNX8 as a novel activator of the sterol regulatory element binding protein (SREBP) transcription factor, a major regulator of cholesterol homeostasis. In that study, we have showed that SNX8 reduced an insulin-induced gene (INSIG)-dependent block of SREBP-mediated transcription. Here, for the first time, we investigated the expression and function of SNX8 within the CNS. We found that SNX8 was expressed within neurons, but not astrocytes or microglia, with neuronal localisation primarily in the soma. The protein levels of neuronal SNX8 were unchanged in the presence of moderately high cholesterol but were decreased by mevinolin (a cholesterol-lowering statin) and U18666a (which causes cholesterol to accumulate within the lysosome). To determine if SNX8 overexpression alters the levels of cholesterol, we engineered a GFP-SNX8 lentivirus. The overexpression of GFP-SNX8 had no effect on cholesterol in neurons under control conditions or in already strongly altered cholesterol conditions of mevinolin or U18666a. In contrast, in moderately high cholesterol, the overexpression of GFP-SNX8 caused redistribution of cholesterol within neurons creating a phenotype similar to U18666a treatment. Taken together, these data suggest that extreme changes in cholesterol reduce SNX8 expression and that overexpression of SNX8 exacerbates aberrant handling of neuronal cholesterol. This work further supports the role for SNX8 in regulating cholesterol levels, which could be important in understanding its role as an Aβ toxicity enhancer and its association with Alzheimer's disease.
- Published
- 2014
- Full Text
- View/download PDF
46. The selective anti-IL17A monoclonal antibody secukinumab (AIN457) attenuates IL17A-induced levels of IL6 in human astrocytes.
- Author
-
Elain G, Jeanneau K, Rutkowska A, Mir AK, and Dev KK
- Subjects
- Animals, Animals, Newborn, Antibodies, Monoclonal, Humanized, Cells, Cultured, Cerebral Cortex drug effects, Cerebral Cortex metabolism, Humans, Male, Mice, Mice, Inbred C57BL, Antibodies, Monoclonal pharmacology, Astrocytes drug effects, Astrocytes metabolism, Interleukin-17 antagonists & inhibitors, Interleukin-17 pharmacology, Interleukin-6 biosynthesis
- Abstract
The family of interleukin 17 receptors (IL17Rs), subtypes IL17RA-IL17RE, is targeted by the group of pro-inflammatory IL17 cytokines (IL17A-F) and moreover the newly developed anti-IL17A antibody secukinumab (AIN457) has shown promise in Phase II trials in multiple sclerosis. Here, we show that human astrocytes, isolated from a fetal cerebral cortex, express IL17RA and IL17RC and in vitro treatment with IL17A increases protein levels of IL6 in human astrocytes, which is enhanced in the presence of TNFα, as determined by homogeneous time resolved fluorescence. Studies on acutely isolated mouse astrocytes are comparable to human astrocytes although the protein levels of IL6 are lower in mouse astrocytes, which also show a lower response to IL17F and IL1β in promoting IL6 levels. In human astrocytes, IL17A and TNFα also induce mRNA expression of IL6, IL8 and the Th17 cytokines CXCL1, CXCL2, and CCL20, with little effect on Th1 cytokines CXCL9, CXCL10, and CXCL11. The effects of IL17A are associated with nuclear translocation of the NF-κB transcription factor, as determined by immunocytochemistry, where treatment of human astrocytes with the inhibitors of the NF-κB pathway and with secukinumab inhibits the IL17A and IL17A/TNFα-induced increase in nuclear translocation of NF-κB and levels of IL6. Taken together the data shows that IL17A signaling plays a key role in regulating the levels of cytokines, such as IL6, in human astrocytes via a mechanism that involves NF-κB signaling and that selective inhibition of IL17A signaling attenuates levels of pro-inflammatory molecules in astrocytes.
- Published
- 2014
- Full Text
- View/download PDF
47. Lymphocyte-mediated neuroprotection in in vitro models of excitotoxicity involves astrocytic activation and the inhibition of MAP kinase signalling pathways.
- Author
-
Shrestha R, Millington O, Brewer J, Dev KK, and Bushell TJ
- Subjects
- Animals, Astrocytes cytology, Astrocytes drug effects, Cell Death drug effects, Cell Death immunology, Culture Media, Conditioned pharmacology, Cytokines immunology, Fluoroacetates toxicity, Hippocampus drug effects, Hippocampus pathology, Lymphocyte Activation immunology, MAP Kinase Signaling System drug effects, Mice, Signal Transduction drug effects, Signal Transduction immunology, Astrocytes immunology, Hippocampus immunology, Kainic Acid toxicity, MAP Kinase Signaling System immunology, Nerve Degeneration chemically induced, Nerve Degeneration immunology, Neuroprotective Agents immunology, T-Lymphocytes immunology
- Abstract
It is well established that immunosurveillance is active in the CNS and plays a key role in several CNS disorders but the exact role of immune cells remains elusive. Thus, in the present study we investigated whether lymphocytes are protective/detrimental in in vitro models of excitotoxicty. Kainate (KA)-induced neuronal death was significantly reduced following exposure to mixed lymphocytes or purified T lymphocytes containing either activated or non-activated T-lymphocytes. Conditioned media from lymphocyte preparations, but not boiled conditioned media, was protective against KA-induced toxicity indicating soluble mediators underlie the observed neuroprotection with cytokine arrays indicating IL-16 as the likely candidate. A role for astrocytes was established as the neuroprotection was abolished in the presence of the glial toxin, fluoroacetate. Furthermore, lymphocytes inhibited p38 MAPK and ERK signalling pathways with pharmacological inhibition of these pathways mimicking the protective effect of lymphocytes. Similarly, lymphocytes were neuroprotective against oxygen-glucose deprivation (OGD)-induced cell death with the inhibition of p38 MAPK and ERK signalling pathways involved. These data indicate that lymphocytes are neuroprotective under our experimental conditions and we suggest that astrocytic activation and inhibition of MAPK signalling cascades are involved but further studies are required to investigate whether similar mechanisms underlie the actions of lymphocytes in in vivo experimental models of disease. This article is part of the Special Issue entitled 'The Synaptic Basis of Neurodegenerative Disorders'., (Copyright © 2013 Elsevier Ltd. All rights reserved.)
- Published
- 2014
- Full Text
- View/download PDF
48. Preanesthesia assessment clinic for cardiac surgery by cardiac anesthesiologist: a practice statement.
- Author
-
Malik M, Panchal AM, and Dev KK
- Subjects
- Counseling, Humans, Informed Consent, Patient Care Team, Referral and Consultation, Anesthesia, Cardiac Surgical Procedures methods, Preoperative Care methods, Thoracic Surgery organization & administration, Thoracic Surgery standards
- Published
- 2014
- Full Text
- View/download PDF
49. Drug dealers: $20 trillion of in-licensing payments.
- Author
-
Frei P and Dev KK
- Subjects
- Humans, Drug Approval, Drug Industry economics, Licensure economics
- Abstract
To help understand the trends of in-licensing, we analysed 781 in-licensing deals costing over $20 trillion over a period of 17 years and found that, while the number of FDA approvals is decreasing, the upfront price paid for drug in-licensing is increasing. Moreover, these upfront payments were not immune to the recent economic downturn, where the yearly increase in upfront payments halted in years 2008-2012. The cost of buying drugs also steadily increased from early research compounds to Phase III drugs. A relative decrease in upfront payments in Filed phase versus to Phase III was seen, which may reflect reluctance in buying filed drugs that are facing the 'patent cliff'. The data here also showed no obvious difference in upfront payments made in five major disease areas, namely, oncology, cardiovascular disease, autoimmunity, nervous system and metabolic disorders., (Copyright © 2013 Elsevier Ltd. All rights reserved.)
- Published
- 2013
- Full Text
- View/download PDF
50. Small-molecule inhibitors at the PSD-95/nNOS interface have antidepressant-like properties in mice.
- Author
-
Doucet MV, Levine H, Dev KK, and Harkin A
- Subjects
- Animals, Antidepressive Agents pharmacology, Chlorophenols pharmacology, Depression metabolism, Disks Large Homolog 4 Protein, Guanylate Kinases metabolism, Hindlimb Suspension methods, Hindlimb Suspension psychology, Male, Membrane Proteins metabolism, Mice, Motor Activity drug effects, Motor Activity physiology, Nitric Oxide Synthase Type I metabolism, Triazoles pharmacology, Antidepressive Agents therapeutic use, Chlorophenols therapeutic use, Depression drug therapy, Guanylate Kinases antagonists & inhibitors, Membrane Proteins antagonists & inhibitors, Nitric Oxide Synthase Type I antagonists & inhibitors, Triazoles therapeutic use
- Abstract
Previous studies have demonstrated that nitric oxide (NO) synthase inhibitors are as efficacious as tricyclic antidepressants in preclinical antidepressant screening procedures and in attenuating behavioural deficits associated with animal models of depression. The N-methyl-D-aspartate receptor (NMDA-R) complex gates Ca(2+), which interacts with calmodulin to subsequently activate NO synthase. We hypothesised that uncoupling neuronal nitric oxide synthase (nNOS) from the NMDA-R through the scaffolding protein postsynaptic density protein 95 (PSD-95) would produce behavioural antidepressant effects similar to NO synthase inhibitors. Small-molecule inhibitors of the PSD-95/nNOS interaction, IC87201 (0.01-2 mg/kg) and ZL006 (10 mg/kg) were tested for antidepressant properties in tests of antidepressant activity namely the tail suspension and forced swim tests in mice. We now report that IC87201 and ZL006 produce antidepressant-like responses in the forced swimming test (FST) and tail suspension test (TST) following a single administration in mice. By contrast to the tricyclic antidepressant imipramine (25 mg/kg), the effects are not observed 1 h following drug administration but are apparent 24 and 72 h later. Furthermore prior exposure to the TST or FST is required in order to observe the antidepressant-related activity. Similar delayed and sustained antidepressant-like effects were observed following TRIM (50 mg/kg) and ketamine (30 mg/kg) in the TST. The antidepressant-like effects of ZL006 also generalise to IC87201 in the TST. IC87201 was devoid of effects on locomotor activity and step-through latency in the passive avoidance cognition test. These data support the hypothesis that targeting the PSD-95/nNOS interaction downstream of NMDA-R produces antidepressant effects and may represent a novel class of therapeutics for major depressive disorders.
- Published
- 2013
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.