Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 71
Filter
Add more filters

Country/Region as subject
Publication year range
1.
J Physiol ; 602(12): 2737-2750, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38795332

ABSTRACT

World Health Organisation data suggest that up to 99% of the global population are exposed to air pollutants above recommended levels. Impacts to health range from increased risk of stroke and cardiovascular disease to chronic respiratory conditions, and air pollution may contribute to over 7 million premature deaths a year. Additionally, mounting evidence suggests that in utero or early life exposure to particulate matter (PM) in ambient air pollution increases the risk of neurodevelopmental impairment with obvious lifelong consequences. Identifying brain-specific cellular targets of PM is vital for determining its long-term consequences. We previously established that microglial-like BV2 cells were particularly sensitive to urban (U)PM-induced damage including reactive oxygen species production, which was abrogated by a mitochondrially targeted antioxidant. Here we extend those studies to find that UPM treatment causes a rapid impairment of mitochondrial function and increased mitochondrial fragmentation. However, there is a subsequent restoration of mitochondrial and therefore cell health occurring concomitantly with upregulated measures of mitochondrial biogenesis and mitochondrial load. Our data highlight that protecting mitochondrial function may represent a valuable mechanism to offset the effects of UPM exposure in the neonatal brain. KEY POINTS: Air pollution represents a growing risk to long-term health especially in early life, and the CNS is emerging a target for airborne particulate matter (PM). We previously showed that microglial-like BV2 cells were vulnerable to urban (U)PM exposure, which impaired cell survival and promoted reactive oxygen species production. Here we find that, following UPM exposure, BV2 mitochondrial membrane potential is rapidly reduced, concomitant with decreased cellular bioenergetics and increased mitochondrial fission. However, markers of mitochondrial biogenesis and mitochondrial mass are subsequently induced, which may represent a cellular mitigation strategy. As mitochondria are more vulnerable in the developing brain, exposure to air pollution may represent a greater risk to lifelong health in this cohort; conversely, promoting mitochondrial integrity may offset these risks.


Subject(s)
Microglia , Mitochondria , Mitochondrial Dynamics , Particulate Matter , Particulate Matter/toxicity , Animals , Mice , Mitochondrial Dynamics/drug effects , Cell Line , Mitochondria/drug effects , Mitochondria/metabolism , Microglia/drug effects , Microglia/metabolism , Organelle Biogenesis , Air Pollutants/toxicity , Reactive Oxygen Species/metabolism
2.
Dev Neurosci ; 2024 Feb 07.
Article in English | MEDLINE | ID: mdl-38325353

ABSTRACT

INTRODUCTION: Sex differences exist in the prevalence of neurodevelopmental disorders (NDDs). Part of the aetiology of NDDs has been proposed to be alterations in the balance between excitatory and inhibitory neurotransmission, leading to the question of whether males and females respond differently to altered neurotransmitter balance. We investigated whether pharmacological alteration of GABAA signalling in early development results in sex-dependent changes in adult behaviours associated with NDDs. METHODS: Male and female C57BL/6J mice received intraperitoneal injections of 0.5mg/kg muscimol or saline on postnatal days (P) 3-5 and were subjected to behavioural testing, specifically open field, light dark box, marble burying, sucralose preference, social interaction and olfactory habituation/dishabituation tests between P60-90. RESULTS: Early postnatal administration of muscimol resulted in reduced anxiety in the light dark box test in both male and female adult mice. Muscimol reduced sucralose preference in males, but not females, whereas female mice showed reduced social behaviours. Regional alterations in cortical thickness were observed in the weeks following GABAA receptor activation, pointing to an evolving structural difference in the brain underlying adult behaviour. CONCLUSIONS: We conclude that activation of the GABAA receptor in the first week of life resulted in long-lasting changes in a range of behaviours in adulthood following altered neurodevelopment. Sex of the individual affected the nature and severity of these abnormalities, explaining part of the varied pathophysiology and neurodevelopmental diagnosis that derive from excitatory/inhibitory imbalance.

3.
Bioessays ; 43(9): e2000288, 2021 09.
Article in English | MEDLINE | ID: mdl-33751627

ABSTRACT

The World Health Organisation recently listed air pollution as the most significant threat to human health. Air pollution comprises particulate matter (PM), metals, black carbon and gases such as ozone (O3 ), nitrogen dioxide (NO2 ) and carbon monoxide (CO). In addition to respiratory and cardiovascular disease, PM exposure is linked with increased risk of neurodegeneration as well as neurodevelopmental impairments. Critically, studies suggest that PM crosses the placenta, making direct in utero exposure a reality. Rodent models reveal that neuroinflammation, neurotransmitter imbalance and oxidative stress are triggered following gestational/early life exposure to PM, and may be exacerbated by concomitant mitochondrial dysfunction. Gestational PM exposure (potentiated by mitochondrial impairment in the metabolically active neonatal brain) not only impacts neurodevelopment but may sensitise the brain to subsequent cognitive impairment. Having reviewed this field, we conclude that strategies are urgently required to reduce exposure to PM during this sensitive developmental period.


Subject(s)
Air Pollutants , Air Pollution , Air Pollutants/analysis , Air Pollutants/toxicity , Air Pollution/adverse effects , Female , Humans , Neuroglia/chemistry , Nitrogen Dioxide/analysis , Particulate Matter/analysis , Particulate Matter/toxicity , Pregnancy
4.
Dev Neurosci ; 44(4-5): 309-319, 2022.
Article in English | MEDLINE | ID: mdl-35500557

ABSTRACT

Air pollution affects the majority of the world's population and has been linked to over 7 million premature deaths per year. Exposure to particulate matter (PM) contained within air pollution is associated with cardiovascular, respiratory, and neurological ill health. There is increasing evidence that exposure to air pollution in utero and in early childhood is associated with altered brain development. However, the underlying mechanisms for impaired brain development are not clear. While oxidative stress and neuroinflammation are documented consequences of PM exposure, cell-specific mechanisms that may be triggered in response to air pollution exposure are less well defined. Here, we assess the effect of urban PM exposure on two different cell types, microglial-like BV2 cells and neural stem/precursor-like C17.2 cells. We found that, contrary to expectations, immature C17.2 cells were more resistant to PM-mediated oxidative stress and cell death than BV2 cells. PM exposure resulted in decreased mitochondrial health and increased mitochondrial ROS in BV2 cells which could be prevented by MitoTEMPO antioxidant treatment. Our data suggest that not only is mitochondrial dysfunction a key trigger in PM-mediated cytotoxicity but that such deleterious effects may also depend on cell type and maturity.


Subject(s)
Air Pollution , Particulate Matter , Air Pollution/adverse effects , Child, Preschool , Humans , Microglia/metabolism , Mitochondria/metabolism , Oxidative Stress , Particulate Matter/toxicity
5.
J Neuroinflammation ; 19(1): 265, 2022 Oct 29.
Article in English | MEDLINE | ID: mdl-36309753

ABSTRACT

Encephalopathy of prematurity (EoP) affects approximately 30% of infants born < 32 weeks gestation and is highly associated with inflammation in the foetus. Here we evaluated the efficacy of montelukast, a cysteinyl leukotriene receptor antagonist widely used to treat asthma in children, to ameliorate peripheral and central inflammation, and subsequent grey matter neuropathology and behaviour deficits in a mouse model of EoP. Male CD-1 mice were treated with intraperitoneal (i.p.) saline or interleukin-1beta (IL-1ß, 40 µg/kg, 5 µL/g body weight) from postnatal day (P)1-5 ± concomitant montelukast (1-30 mg/kg). Saline or montelukast treatment was continued for a further 5 days post-injury. Assessment of systemic and central inflammation and short-term neuropathology was performed from 4 h following treatment through to P10. Behavioural testing, MRI and neuropathological assessments were made on a second cohort of animals from P36 to 54. Montelukast was found to attenuate both peripheral and central inflammation, reducing the expression of pro-inflammatory molecules (IL-1ß, IL-6, TNF) in the brain. Inflammation induced a reduction in parvalbumin-positive interneuron density in the cortex, which was normalised with high-dose montelukast. The lowest effective dose, 3 mg/kg, was able to improve anxiety and spatial learning deficits in this model of inflammatory injury, and alterations in cortical mean diffusivity were not present in animals that received this dose of montelukast. Repurposed montelukast administered early after preterm birth may, therefore, improve grey matter development and outcome in EoP.


Subject(s)
Brain Diseases , Premature Birth , Quinolines , Infant, Newborn , Humans , Female , Male , Animals , Mice , Gray Matter , Premature Birth/drug therapy , Acetates/therapeutic use , Acetates/pharmacology , Quinolines/therapeutic use , Quinolines/pharmacology , Disease Models, Animal , Inflammation/drug therapy
6.
Biochem J ; 478(4): 749-764, 2021 02 26.
Article in English | MEDLINE | ID: mdl-33626141

ABSTRACT

The incidence of cocaine abuse is increasing especially in the U.K. where the rates are among the highest in Europe. In addition to its role as a psychostimulant, cocaine has profound effect on brain metabolism, impacting glycolysis and impairing oxidative phosphorylation. Cocaine exposure alters metabolic gene expression and protein networks in brain regions including the prefrontal cortex, the ventral tegmental area and the nucleus accumbens, the principal nuclei of the brain reward system. Here, we focus on how cocaine impacts mitochondrial function, in particular through alterations in electron transport chain function, reactive oxygen species (ROS) production and oxidative stress (OS), mitochondrial dynamics and mitophagy. Finally, we describe the impact of cocaine on brain energy metabolism in the developing brain following prenatal exposure. The plethora of mitochondrial functions altered following cocaine exposure suggest that therapies maintaining mitochondrial functional integrity may hold promise in mitigating cocaine pathology and addiction.


Subject(s)
Cocaine-Related Disorders/metabolism , Mitochondria/physiology , Animals , Antioxidants/pharmacology , Antioxidants/therapeutic use , Brain/drug effects , Brain/embryology , Brain/metabolism , Cocaine/pharmacology , Cocaine/toxicity , Energy Metabolism/drug effects , Female , Glycolysis/drug effects , Humans , Mice , Mitochondrial Transmembrane Permeability-Driven Necrosis/drug effects , Mitochondrial Turnover/drug effects , Neuroglia/drug effects , Neuroglia/metabolism , Organelle Biogenesis , Oxidative Phosphorylation/drug effects , Oxidative Stress , Pregnancy , Pregnancy Complications/chemically induced , Prenatal Exposure Delayed Effects , Rats , Reactive Oxygen Species , Reward , Substance Withdrawal Syndrome/metabolism
7.
Glia ; 67(6): 1047-1061, 2019 06.
Article in English | MEDLINE | ID: mdl-30637805

ABSTRACT

Accumulating evidence suggests that changes in the metabolic signature of microglia underlie their response to inflammation. We sought to increase our knowledge of how pro-inflammatory stimuli induce metabolic changes. Primary microglia exposed to lipopolysaccharide (LPS)-expressed excessive fission leading to more fragmented mitochondria than tubular mitochondria. LPS-mediated Toll-like receptor 4 (TLR4) activation also resulted in metabolic reprogramming from oxidative phosphorylation to glycolysis. Blockade of mitochondrial fission by Mdivi-1, a putative mitochondrial division inhibitor led to the reversal of the metabolic shift. Mdivi-1 treatment also normalized the changes caused by LPS exposure, namely an increase in mitochondrial reactive oxygen species production and mitochondrial membrane potential as well as accumulation of key metabolic intermediate of TCA cycle succinate. Moreover, Mdivi-1 treatment substantially reduced LPS induced cytokine and chemokine production. Finally, we showed that Mdivi-1 treatment attenuated expression of genes related to cytotoxic, repair, and immunomodulatory microglia phenotypes in an in vivo neuroinflammation paradigm. Collectively, our data show that the activation of microglia to a classically pro-inflammatory state is associated with a switch to glycolysis that is mediated by mitochondrial fission, a process which may be a pharmacological target for immunomodulation.


Subject(s)
Energy Metabolism/drug effects , Inflammation Mediators/metabolism , Lipopolysaccharides/toxicity , Microglia/drug effects , Microglia/metabolism , Reactive Oxygen Species/metabolism , Animals , Energy Metabolism/physiology , Female , Inflammation/chemically induced , Inflammation/metabolism , Inflammation/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microglia/pathology , Oxidative Phosphorylation/drug effects , Pregnancy
8.
Brain ; 141(10): 2925-2942, 2018 10 01.
Article in English | MEDLINE | ID: mdl-30165597

ABSTRACT

Hypoxic-ischaemic encephalopathy remains a global health burden. Despite medical advances and treatment with therapeutic hypothermia, over 50% of cooled infants are not protected and still develop lifelong neurodisabilities, including cerebral palsy. Furthermore, hypothermia is not used in preterm cases or low resource settings. Alternatives or adjunct therapies are urgently needed. Exendin-4 is a drug used to treat type 2 diabetes mellitus that has also demonstrated neuroprotective properties, and is currently being tested in clinical trials for Alzheimer's and Parkinson's diseases. Therefore, we hypothesized a neuroprotective effect for exendin-4 in neonatal neurodisorders, particularly in the treatment of neonatal hypoxic-ischaemic encephalopathy. Initially, we confirmed that the glucagon like peptide 1 receptor (GLP1R) was expressed in the human neonatal brain and in murine neurons at postnatal Day 7 (human equivalent late preterm) and postnatal Day 10 (term). Using a well characterized mouse model of neonatal hypoxic-ischaemic brain injury, we investigated the potential neuroprotective effect of exendin-4 in both postnatal Day 7 and 10 mice. An optimal exendin-4 treatment dosing regimen was identified, where four high doses (0.5 µg/g) starting at 0 h, then at 12 h, 24 h and 36 h after postnatal Day 7 hypoxic-ischaemic insult resulted in significant brain neuroprotection. Furthermore, neuroprotection was sustained even when treatment using exendin-4 was delayed by 2 h post hypoxic-ischaemic brain injury. This protective effect was observed in various histopathological markers: tissue infarction, cell death, astrogliosis, microglial and endothelial activation. Blood glucose levels were not altered by high dose exendin-4 administration when compared to controls. Exendin-4 administration did not result in adverse organ histopathology (haematoxylin and eosin) or inflammation (CD68). Despite initial reduced weight gain, animals restored weight gain following end of treatment. Overall high dose exendin-4 administration was well tolerated. To mimic the clinical scenario, postnatal Day 10 mice underwent exendin-4 and therapeutic hypothermia treatment, either alone or in combination, and brain tissue loss was assessed after 1 week. Exendin-4 treatment resulted in significant neuroprotection alone, and enhanced the cerebroprotective effect of therapeutic hypothermia. In summary, the safety and tolerance of high dose exendin-4 administrations, combined with its neuroprotective effect alone or in conjunction with clinically relevant hypothermia make the repurposing of exendin-4 for the treatment of neonatal hypoxic-ischaemic encephalopathy particularly promising.


Subject(s)
Brain/drug effects , Exenatide/pharmacology , Hypoxia-Ischemia, Brain/pathology , Neuroprotective Agents/pharmacology , Animals , Animals, Newborn , Brain/pathology , Disease Models, Animal , Humans , Hypothermia, Induced , Mice
9.
Dev Neurosci ; 39(1-4): 192-206, 2017.
Article in English | MEDLINE | ID: mdl-28402971

ABSTRACT

Inflammation in the perinatal brain caused by maternal or intrauterine fetal infection is now well established as an important contributor to the development of perinatal brain injury. Exposure to inflammatory products can impair perinatal brain development and act as a risk factor for neurological dysfunction, cognitive disorders, cerebral palsy, or preterm birth. Pre-exposure to inflammation significantly exacerbates brain injury caused by hypoxic/ischaemic insult. Tumour necrosis factor (TNF) is a family of cytokines largely involved in inflammation signalling. In our previous study, we identified the importance of TNF-related apoptosis-inducing ligand (TRAIL) signalling in the development of perinatal brain injury. We observed a significant increase in the expression levels of a soluble decoy receptor for TRAIL, osteoprotegerin (OPG). Besides TRAIL, OPG is able to bind the receptor activator of the NF-κB (RANK) ligand (RANKL) and inhibit its signalling. The function of the RANK/RANKL/OPG system in the brain has not come under much scrutiny. The aim of this research study was to elucidate the role of RANK, RANKL, and OPG in microglial responses to the proinflammatory stimuli lipopolysaccharide (LPS) and polyinosinic-polycytidylic acid (Poly I:C). Here, we show that RANK signalling is important for regulating the activation of the BV2 microglial cell line. We found that LPS treatment causes a significant decrease in the expression of RANK in the BV2 cell line while significantly increasing the expression of OPG, Toll-like receptor (TLR)3, and the adaptor proteins MyD88 and TRIF. We found that pretreatment of BV2 cells with RANKL for 24 h before the LPS or Poly I:C exposure decreases the expression of inflammatory markers such as inducible nitric oxide synthase and cyclooxygenase. This is accompanied by a decreased expression of the TLR adaptor proteins MyD88 and TRIF, which we observed after RANKL treatment. Similar results were obtained in our experiments with primary mouse microglia. Using recently developed CRISPR/Cas9 technology, we generated a BV2 cell line lacking RANK (RANK-/- BV2). We showed that most effects of RANKL pretreatment were abolished, thereby proving the specificity of this effect. Taken together, these findings suggest that RANK signalling is important for modulating the inflammatory activation of microglial cells to a moderate level, and that RANK attenuates TLR3/TLR4 signalling.


Subject(s)
Brain Diseases/metabolism , Microglia/metabolism , NF-kappa B/metabolism , RANK Ligand/metabolism , Toll-Like Receptors/metabolism , Animals , Animals, Newborn , Brain Diseases/immunology , Cell Line , Inflammation/chemically induced , Inflammation/immunology , Inflammation/metabolism , Lipopolysaccharides/toxicity , Microglia/immunology , NF-kappa B/immunology , Poly I-C/toxicity , RANK Ligand/immunology , Rats , Rats, Sprague-Dawley , Signal Transduction/physiology , Toll-Like Receptors/immunology
10.
J Neuroinflammation ; 14(1): 255, 2017 Dec 20.
Article in English | MEDLINE | ID: mdl-29262837

ABSTRACT

BACKGROUND: Infection and sepsis are associated with brain white matter injury in preterm infants and the subsequent development of cerebral palsy. METHODS: In the present study, we used a neonatal mouse sepsis-induced white matter injury model to determine the contribution of different T cell subsets (αßT cells and γδT cells) to white matter injury and consequent behavioral changes. C57BL/6J wild-type (WT), T cell receptor (TCR) δ-deficient (Tcrd -/-, lacking γδT cells), and TCRα-deficient (Tcra -/-, lacking αßT cells) mice were administered with lipopolysaccharide (LPS) at postnatal day (PND) 2. Brain myelination was examined at PNDs 12, 26, and 60. Motor function and anxiety-like behavior were evaluated at PND 26 or 30 using DigiGait analysis and an elevated plus maze. RESULTS: White matter development was normal in Tcrd -/- and Tcrα -/- compared to WT mice. LPS exposure induced reductions in white matter tissue volume in WT and Tcrα -/- mice, but not in the Tcrd -/- mice, compared with the saline-treated groups. Neither LPS administration nor the T cell deficiency affected anxiety behavior in these mice as determined with the elevated plus maze. DigiGait analysis revealed motor function deficiency after LPS-induced sepsis in both WT and Tcrα -/- mice, but no such effect was observed in Tcrd -/- mice. CONCLUSIONS: Our results suggest that γδT cells but not αßT cells contribute to sepsis-induced white matter injury and subsequent motor function abnormalities in early life. Modulating the activity of γδT cells in the early stages of preterm white matter injury might represent a novel therapeutic strategy for the treatment of perinatal brain injury.


Subject(s)
Leukoencephalopathies/etiology , Movement Disorders/etiology , Receptors, Antigen, T-Cell, alpha-beta/deficiency , Receptors, Antigen, T-Cell, gamma-delta/deficiency , Sepsis/complications , Animals , Animals, Newborn , Anxiety/etiology , Anxiety/genetics , Brain/drug effects , Brain/growth & development , Brain/metabolism , Brain/pathology , Cytokines/metabolism , Disease Models, Animal , Gait/drug effects , Gait/genetics , Gene Expression Regulation, Developmental/drug effects , Gene Expression Regulation, Developmental/genetics , Lipopolysaccharides/toxicity , Mice , Mice, Inbred C57BL , Mice, Transgenic , Myelin Basic Protein/metabolism , Receptors, Antigen, T-Cell, alpha-beta/genetics , Receptors, Antigen, T-Cell, gamma-delta/genetics , Sepsis/chemically induced , Sepsis/pathology , Spleen/pathology , T-Lymphocyte Subsets/metabolism , T-Lymphocyte Subsets/pathology
11.
Biochem Soc Trans ; 45(5): 1067-1076, 2017 Oct 15.
Article in English | MEDLINE | ID: mdl-28939695

ABSTRACT

Birth asphyxia in term neonates affects 1-2/1000 live births and results in the development of hypoxic-ischaemic encephalopathy with devastating life-long consequences. The majority of neuronal cell death occurs with a delay, providing the potential of a treatment window within which to act. Currently, treatment options are limited to therapeutic hypothermia which is not universally successful. To identify new interventions, we need to understand the molecular mechanisms underlying the injury. Here, we provide an overview of the contribution of both oxidative stress and endoplasmic reticulum stress in the development of neonatal brain injury and identify current preclinical therapeutic strategies.


Subject(s)
Asphyxia Neonatorum/complications , Hypoxia-Ischemia, Brain/etiology , Reactive Oxygen Species/metabolism , Animals , Asphyxia Neonatorum/drug therapy , Asphyxia Neonatorum/metabolism , Endoplasmic Reticulum Stress/drug effects , Humans , Hypoxia-Ischemia, Brain/drug therapy , Hypoxia-Ischemia, Brain/metabolism , Infant, Newborn , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , Oxidative Stress/drug effects
12.
J Biol Chem ; 289(13): 9430-9, 2014 Mar 28.
Article in English | MEDLINE | ID: mdl-24509861

ABSTRACT

Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) is a member of the TNF family. The interaction of TRAIL with death receptor 4 (DR4) and DR5 can trigger apoptotic cell death. The aim of this study was to investigate the role of TRAIL signaling in neonatal hypoxia-ischemia (HI). Using a neonatal mouse model of HI, mRNA, and protein expression of TRAIL, DR5 and the TRAIL decoy receptors osteoprotegerin (OPG), mDcTRAILR1, and mDcTRAILR2 were determined. In vitro, mRNA expression of these genes was measured in primary neurons and oligodendrocyte progenitor cells (OPCs) after inflammatory cytokine (TNF-α/IFN-γ) treatment and/or oxygen and glucose deprivation (OGD). The toxicity of these various paradigms was also measured. The expression of TRAIL, DR5, OPG, and mDcTRAILR2 was significantly increased after HI. In vitro, inflammatory cytokines and OGD treatment significantly induced mRNAs for TRAIL, DR5, OPG, and mDcTRAILR2 in primary neurons and of TRAIL and OPG in OPCs. TRAIL protein was expressed primarily in microglia and astroglia, whereas DR5 co-localized with neurons and OPCs in vivo. OGD enhanced TNF-α/IFN-γ toxicity in both neuronal and OPC cultures. Recombinant TRAIL exerted toxicity alone or in combination with OGD and TNF-α/IFN-γ in primary neurons but not in OPC cultures. The marked increases in the expression of TRAIL and its receptors after cytokine exposure and OGD in primary neurons and OPCs were similar to those found in our animal model of neonatal HI. The toxicity of TRAIL in primary neurons suggests that TRAIL signaling participates in neonatal brain injury after inflammation and HI.


Subject(s)
Central Nervous System/pathology , Hypoxia/pathology , Ischemia/pathology , Signal Transduction , TNF-Related Apoptosis-Inducing Ligand/metabolism , Animals , Cell Death/drug effects , Female , Gene Expression Regulation/drug effects , Glucose/deficiency , Hypoxia/metabolism , Inflammation/metabolism , Inflammation/pathology , Interferon-gamma/pharmacology , Ischemia/metabolism , Male , Mice , Mice, Inbred C57BL , Neurons/metabolism , Neurons/pathology , Oligodendroglia/pathology , Oxygen/metabolism , Receptors, TNF-Related Apoptosis-Inducing Ligand/metabolism , Signal Transduction/drug effects , Stem Cells/drug effects , Stem Cells/pathology , TNF-Related Apoptosis-Inducing Ligand/genetics , Tumor Necrosis Factor-alpha/pharmacology
13.
J Neurochem ; 133(2): 242-52, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25598140

ABSTRACT

Perinatal hypoxic-ischaemic encephalopathy (HIE) occurs in 1-2 in every 1000 term infants and the devastating consequences range from cerebral palsy, epilepsy and neurological deficit to death. Cellular damage post insult occurs after a delay and is mediated by a secondary neural energy failure. AMP-activated protein kinase (AMPK) is a sensor of cellular stress resulting from ATP depletion and/or calcium dysregulation, hallmarks of the neuronal cell death observed after HIE. AMPK activation has been implicated in the models of adult ischaemic injury but, as yet, there have been no studies defining its role in neonatal asphyxia. Here, we find that in an in vivo model of neonatal hypoxia-ischaemic and in oxygen/glucose deprivation in neurons, there is pathological activation of the calcium/calmodulin-dependent protein kinase kinase ß (CaMKKß)-AMPKα1 signalling pathway. Pharmacological inhibition of AMPK during the insult promotes neuronal survival but, conversely, inhibiting AMPK activity prior to the insult sensitizes neurons, exacerbating cell death. Our data have pathological relevance for neonatal HIE as prior sensitization such as exposure to bacterial infection (reported to reduce AMPK activity) produces a significant increase in injury. We show that in an in vivo model of neonatal hypoxia-ischaemic and in oxygen/glucose deprivation in neurons, there is a pathological activation of the CaMKKß-AMPKα1 signalling pathway. Inhibiting AMPK during OGD promotes neuronal survival; conversely, inhibiting AMPK prior to OGD exacerbates cell death. Our data have clinical relevance as prior sensitization (e.g. exposure to bacterial infection reducing AMPK activity) increases injury. AMPK, AMP-activated protein kinase; HI, hypoxia-ischaemia; OGD, oxygen-glucose deprivation.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Brain/metabolism , Gene Expression Regulation, Developmental/physiology , Hypoxia-Ischemia, Brain/enzymology , Hypoxia-Ischemia, Brain/pathology , Animals , Animals, Newborn , Benzimidazoles/pharmacology , Cell Death , Cells, Cultured , Disease Models, Animal , Enzyme Inhibitors/pharmacology , Glucose/deficiency , Hypoxia/pathology , Ionomycin/pharmacology , L-Lactate Dehydrogenase/metabolism , Mice , Mice, Inbred C57BL , Naphthalimides/pharmacology , Neurons/metabolism , Signal Transduction/physiology , Time Factors
14.
Dev Neurosci ; 37(4-5): 363-75, 2015.
Article in English | MEDLINE | ID: mdl-25721106

ABSTRACT

Neonatal encephalopathy (NE) is a leading cause of childhood death and disability in term infants. Treatment options for perinatal brain injury are limited and developing therapies that target multiple pathways within the pathophysiology of NE are of great interest. Pifithrin-µ (PFT-µ) is a drug with striking neuroprotective abilities in a preclinical model of hypoxia-ischemia (HI)-induced NE wherein cell death is a substantial cause of injury. Work from neurons and tumor cells reports that PFT-µ is able to inhibit p53 binding to the mitochondria, heat shock protein (HSP)-70 substrate binding and activation of the NF-kB pathway. The purpose of this study is to understand whether the neuroprotective effects of PFT-µ also include direct effects on microglia. We utilized the microglial cell line, BV2, and we studied the dose-dependent effect of PFT-µ on M1-like and M2-like phenotype using qRT-PCR and Western blotting, including the requirement for the presence of p53 or HSP-70 in these effects. We also assessed phagocytosis and the effects of PFT-µ on genes within metabolic pathways related to phenotype. We noted that PFT-µ robustly reduced the M1-like (lipopolysaccharide, LPS-induced) BV2 response, spared the LPS-induced phagocytic ability of BV2 and had no effect on the genes related to metabolism and that effects on phenotype were partially dependent on the presence of HSP-70 but not p53. This study demonstrates that the neuroprotective effects of PFT-µ in HI-induced NE may include an anti-inflammatory effect on microglia and adds to the evidence that this drug might be of clinical interest for the treatment of NE.


Subject(s)
Benzothiazoles/pharmacology , Gene Expression/drug effects , Inflammation/drug therapy , Microglia/drug effects , Neuroprotective Agents/pharmacology , Toluene/analogs & derivatives , Tumor Suppressor Protein p53/metabolism , Animals , Cell Line , Inflammation/immunology , Mice , Microglia/immunology , Mitochondria/drug effects , Mitochondria/immunology , Toluene/pharmacology , Tumor Suppressor Protein p53/antagonists & inhibitors
15.
Dev Neurosci ; 37(4-5): 321-37, 2015.
Article in English | MEDLINE | ID: mdl-25823427

ABSTRACT

Apoptotic mechanisms are centre stage for the development of injury in the immature brain, and caspases have been shown to play a pivotal role during brain development and in response to injury. The inhibition of caspases using broad-spectrum agents such as Q-VD-OPh is neuroprotective in the immature brain. Caspase-6, an effector caspase, has been widely researched in neurodevelopmental disorders and found to be important following adult stroke, but its function in the neonatal brain has yet to be detailed. Furthermore, caspases may be important in microglial activation; microglia are required for optimal brain development and following injury, and their close involvement during neuronal cell death suggests that apoptotic cues such as caspase activation may be important in microglial activation. Therefore, in this study we aimed to investigate the possible apoptotic and non-apoptotic functions caspase-6 may have in the immature brain in response to hypoxia-ischaemia. We examined whether caspases are involved in microglial activation. We assessed cleaved caspase-6 expression following hypoxia-ischaemia and conducted primary microglial cultures to assess whether the broad-spectrum inhibitor Q-VD-OPh or caspase-6 gene deletion affected lipopolysaccharide (LPS)-mediated microglial activation and phenotype. We observed cleaved caspase-6 expression to be low but present in the cell body and cell processes in both a human case of white matter injury and 72 h following hypoxia-ischaemia in the rat. Gene deletion of caspase-6 did not affect the outcome of brain injury following mild (50 min) or severe (60 min) hypoxia-ischaemia. Interestingly, we did note that cleaved caspase-6 was co-localised with microglia that were not of apoptotic morphology. We observed that mRNA of a number of caspases was modulated by low-dose LPS stimulation of primary microglia. Q-VD-OPh treatment and caspase-6 gene deletion did not affect microglial activation but modified slightly the M2b phenotype response by changing the time course of SOCS3 expression after LPS administration. Our results suggest that the impact of active caspase-6 in the developing brain is subtle, and we believe there are predominantly other caspases (caspase-2, -3, -8, -9) that are essential for the cell death processes in the immature brain.


Subject(s)
Brain Injuries/metabolism , Brain/growth & development , Caspase 6/metabolism , Hypoxia-Ischemia, Brain/metabolism , Microglia/metabolism , Animals , Animals, Newborn , Brain/metabolism , Brain Injuries/etiology , Hypoxia-Ischemia, Brain/complications , Mice , Mice, Knockout , Rats
16.
Int J Mol Sci ; 16(9): 22509-26, 2015 Sep 17.
Article in English | MEDLINE | ID: mdl-26393574

ABSTRACT

Perturbation of mitochondrial function and subsequent induction of cell death pathways are key hallmarks in neonatal hypoxic-ischemic (HI) injury, both in animal models and in term infants. Mitoprotective therapies therefore offer a new avenue for intervention for the babies who suffer life-long disabilities as a result of birth asphyxia. Here we show that after oxygen-glucose deprivation in primary neurons or in a mouse model of HI, mitochondrial protein homeostasis is altered, manifesting as a change in mitochondrial morphology and functional impairment. Furthermore we find that the mitochondrial fusion and cristae regulatory protein, OPA1, is aberrantly cleaved to shorter forms. OPA1 cleavage is normally regulated by a balanced action of the proteases Yme1L and Oma1. However, in primary neurons or after HI in vivo, protein expression of YmelL is also reduced, whereas no change is observed in Oma1 expression. Our data strongly suggest that alterations in mitochondria-shaping proteins are an early event in the pathogenesis of neonatal HI injury.


Subject(s)
GTP Phosphohydrolases/metabolism , Hypoxia-Ischemia, Brain/metabolism , Metalloendopeptidases/metabolism , Neurons/metabolism , Animals , Animals, Newborn , Cell Hypoxia , Cell Line , Disease Models, Animal , Gene Expression Regulation, Developmental , Hypoxia-Ischemia, Brain/pathology , Membrane Potential, Mitochondrial , Metalloproteases/metabolism , Mice , Mitochondria/pathology , Mitochondria/physiology , Mitochondrial Proteins/metabolism , Protein Processing, Post-Translational
17.
FASEB J ; 27(9): 3797-804, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23737250

ABSTRACT

Complement is an essential component of inflammation that plays a role in ischemic brain injury. Recent reports demonstrate novel functions of complement in normal and diseased CNS, such as regulation of neurogenesis and synapse elimination. Here, we examined the role of complement-derived peptide C3a in unilateral hypoxia-ischemia (HI), a model of neonatal HI encephalopathy. HI injury was induced at postnatal day 9 (P9), and loss of hippocampal tissue was determined on P31. We compared WT mice with transgenic mice expressing C3a under the control of glial fibrillary acidic protein promoter, which express biologically active C3a only in CNS and without the requirement of a priori complement activation. Further, we injected C3a peptide into the lateral cerebral ventricle of mice lacking the C3a receptor (C3aR) and WT mice and assessed HI-induced memory impairment 41 d later. We found that HI-induced tissue loss in C3a overexpressing mice was reduced by 50% compared with WT mice. C3a peptide injected 1 h after HI protected WT but not C3aR-deficient mice against HI-induced memory impairment. Thus, C3a acting through its canonical receptor ameliorates behavioral deficits after HI injury, and C3aR is a novel therapeutic target for the treatment of neonatal HI encephalopathy.


Subject(s)
Hypoxia-Ischemia, Brain/metabolism , Receptors, Complement/metabolism , Animals , Animals, Newborn , Astrocytes/cytology , Astrocytes/metabolism , Complement C3a/metabolism , Hippocampus/metabolism , Hippocampus/pathology , Hypoxia-Ischemia, Brain/genetics , Hypoxia-Ischemia, Brain/pathology , Immunohistochemistry , Macrophages/cytology , Macrophages/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neurons/cytology , Neurons/metabolism , RNA, Messenger/genetics , Receptors, Complement/deficiency , Receptors, Complement/genetics
18.
J Pain ; 25(6): 104450, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38154623

ABSTRACT

The evidence that athletes respond to and report indices of experimental pain differently to non-athlete populations was analysed. Databases screened were SPORTDiscus, PubMED, PsycArticles, the Cochrane Library (Cochrane Database of Systematic Reviews), Web of Science, Scopus, and CINAHL. Studies that compared experimentally induced pain responses (threshold, tolerance, intensity, unpleasantness, bothersomeness, and effect on performance) in athletes and controls were included. Meta-analyses were performed where appropriate and effects were described as standardised mean differences, pooled using random effects models. Thirty-six studies (2,492 participants) met the inclusion criteria comprising 19 pain tolerance, 17 pain threshold, 21 pain intensity, 5 pain unpleasantness, 2 performance in pain and 1 bothersomeness study. Athletes demonstrated greater pain tolerance (g = .88 [95% confidence interval [CI] .65, .13]) and reported less pain intensity (g = -.80, [95% CI -1.13, -.47]) compared to controls; they also had higher pain threshold but with smaller effects (g = .41, [95% CI .08, .75]). Differences for unpleasantness did not reach statistical significance but the effects were large (g = -1.23 [95% CI -2.29, .18]). Two studies reported that performance in pain was better in contact athletes than non-athletes, and one concluded that athletes find pain less bothersome than controls. There were considerable inconsistencies in the methods employed that were reflected in the meta-analyses' findings. Sub-group analyses of tolerance and intensity were conducted between endurance, contact, and other athlete groups, but were not significant. The data suggest that athletic participation is associated with altered pain responses, but mechanisms remain unclear and more transparent methods are recommended.This study was registered on the PROSPERO site in January 2019 (ref ID: CRD42019119611). PERSPECTIVE: This review examined differences in pain outcomes (threshold, tolerance, intensity, unpleasantness, bothersomeness) and the effect of pain on performance, in athletes versus controls. Meta-analyses revealed athletes had higher threshold and tolerance and found pain less intense than controls; there was some evidence of differences in bothersomeness and performance.


Subject(s)
Athletes , Humans , Athletes/psychology , Pain Threshold/physiology , Pain/physiopathology , Pain/psychology
19.
Cells ; 13(7)2024 Apr 04.
Article in English | MEDLINE | ID: mdl-38607070

ABSTRACT

Neonatal hypoxia-ischemia (HI) affects 2-3 per 1000 live births in developed countries and up to 26 per 1000 live births in developing countries. It is estimated that of the 750,000 infants experiencing a hypoxic-ischemic event during birth per year, more than 400,000 will be severely affected. As treatment options are limited, rapidly identifying new therapeutic avenues is critical, and repurposing drugs already in clinical use offers a fast-track route to clinic. One emerging avenue for therapeutic intervention in neonatal HI is to target mitochondrial dysfunction, which occurs early in the development of brain injury. Mitochondrial dynamics are particularly affected, with mitochondrial fragmentation occurring at the expense of the pro-fusion protein Optic Atrophy (OPA)1. OPA1, together with mitofusins (MFN)1/2, are required for membrane fusion, and therefore, protecting their function may also safeguard mitochondrial dynamics. Leflunomide, an FDA-approved immunosuppressant, was recently identified as an activator of MFN2 with partial effects on OPA1 expression. We, therefore, treated C17.2 cells with Leflunomide before or after oxygen-glucose deprivation, an in vitro mimic of HI, to determine its efficacy as a neuroprotection and inhibitor of mitochondrial dysfunction. Leflunomide increased baseline OPA1 but not MFN2 expression in C17.2 cells. However, Leflunomide was unable to promote cell survival following OGD. Equally, there was no obvious effect on mitochondrial morphology or bioenergetics. These data align with studies suggesting that the tissue and mitochondrial protein profile of the target cell/tissue are critical for taking advantage of the therapeutic actions of Leflunomide.


Subject(s)
Mitochondrial Diseases , Oxygen , Infant, Newborn , Humans , Oxygen/metabolism , Glucose/metabolism , Leflunomide/pharmacology , Cells, Cultured
20.
EMBO Mol Med ; 16(6): 1284-1309, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38783166

ABSTRACT

Hypoxic-ischaemic encephalopathy (HIE) arises from diminished blood flow and oxygen to the neonatal brain during labor, leading to infant mortality or severe brain damage, with a global incidence of 1.5 per 1000 live births. Glucagon-like Peptide 1 Receptor (GLP1-R) agonists, used in type 2 diabetes treatment, exhibit neuroprotective effects in various brain injury models, including HIE. In this study, we observed enhanced neurological outcomes in post-natal day 10 mice with surgically induced hypoxic-ischaemic (HI) brain injury after immediate systemic administration of exendin-4 or semaglutide. Short- and long-term assessments revealed improved neuropathology, survival rates, and locomotor function. We explored the mechanisms by which GLP1-R agonists trigger neuroprotection and reduce inflammation following oxygen-glucose deprivation and HI in neonatal mice, highlighting the upregulation of the PI3/AKT signalling pathway and increased cAMP levels. These findings shed light on the neuroprotective and anti-inflammatory effects of GLP1-R agonists in HIE, potentially extending to other neurological conditions, supporting their potential clinical use in treating infants with HIE.


Subject(s)
Animals, Newborn , Disease Models, Animal , Hypoxia-Ischemia, Brain , Neuroprotective Agents , Animals , Hypoxia-Ischemia, Brain/drug therapy , Hypoxia-Ischemia, Brain/metabolism , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , Mice , Signal Transduction/drug effects , Exenatide/pharmacology , Exenatide/therapeutic use , Hypoglycemic Agents/pharmacology , Glucagon-Like Peptide-1 Receptor/metabolism , Glucagon-Like Peptide-1 Receptor/agonists , Peptides/pharmacology , Peptides/therapeutic use
SELECTION OF CITATIONS
SEARCH DETAIL