Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 44
Filter
Add more filters










Publication year range
2.
Hum Mol Genet ; 31(8): 1242-1262, 2022 04 22.
Article in English | MEDLINE | ID: mdl-34718572

ABSTRACT

The regeneration-associated gene (RAG) expression program is activated in injured peripheral neurons after axotomy and enables long-distance axon re-growth. Over 1000 genes are regulated, and many transcription factors are upregulated or activated as part of this response. However, a detailed picture of how RAG expression is regulated is lacking. In particular, the transcriptional targets and specific functions of the various transcription factors are unclear. Jun was the first-regeneration-associated transcription factor identified and the first shown to be functionally important. Here we fully define the role of Jun in the RAG expression program in regenerating facial motor neurons. At 1, 4 and 14 days after axotomy, Jun upregulates 11, 23 and 44% of the RAG program, respectively. Jun functions relevant to regeneration include cytoskeleton production, metabolic functions and cell activation, and the downregulation of neurotransmission machinery. In silico analysis of promoter regions of Jun targets identifies stronger over-representation of AP1-like sites than CRE-like sites, although CRE sites were also over-represented in regions flanking AP1 sites. Strikingly, in motor neurons lacking Jun, an alternative SRF-dependent gene expression program is initiated after axotomy. The promoters of these newly expressed genes exhibit over-representation of CRE sites in regions near to SRF target sites. This alternative gene expression program includes plasticity-associated transcription factors and leads to an aberrant early increase in synapse density on motor neurons. Jun thus has the important function in the early phase after axotomy of pushing the injured neuron away from a plasticity response and towards a regenerative phenotype.


Subject(s)
Axons , Nerve Regeneration , Axons/metabolism , Axotomy , Motor Neurons/metabolism , Nerve Regeneration/genetics , Proto-Oncogene Proteins c-jun/genetics , Proto-Oncogene Proteins c-jun/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism
3.
J Physiol ; 596(23): 6043-6062, 2018 12.
Article in English | MEDLINE | ID: mdl-29873394

ABSTRACT

KEY POINTS: This study identifies phosphorylated extracellular signal-regulated kinase (ERK) to be immediately diminished followed by a rapid if transient increase for up to 4 h following hypoxic-ischaemic insult (HI) in the neonatal mouse. Phosphorylated ERK up-regulation was prevented with systemic injection of the mitogen-activated protein kinase kinase (MEK) inhibitor SL327. Treatment with SL327 both pre- and post-HI gave a strong reduction in the number of dying cells and microgliosis. By utilising transgenic mouse mutations, we observe that neuronal ERK2 significantly contributes to tissue damage, while ERK1 and astrocytic ERK2 are neuroprotective. Compared to global inactivation, selective cell-specific interference with ERK activity could result in stronger neuroprotection. ABSTRACT: Hypoxia-ischaemia (HI) is a major cause of neonatal brain injury resulting in cerebral palsy, epilepsy, cognitive impairment and other neurological disabilities. The role of extracellular signal-regulated kinase (ERK) isoforms and their mitogen-activated protein kinase kinase (MEK)-dependent phosphorylation in HI has previously been explored but remains unresolved at cellular level. This is pertinent given the growing awareness of the role of non-neuronal cells in neuroprotection. Using a modified Rice-Vannucci model of HI in the neonatal mouse we observed time- and cell-dependent ERK phosphorylation (pERK), with strongly up-regulated pERK immunoreactivity first in periventricular white matter axons within 15-45 min of HI, followed by forebrain astrocytes and neurons (1-4 h post-HI), and return to baseline by 16 h. We explored the effects of pharmacological ERK blockade through the MEK inhibitor SL327 on neonatal HI-brain damage following HI alone (30 or 60 min) or lipopolysaccharide (LPS)-sensitised HI insult (30 min). Global inhibition of ERK phosphorylation with systemically applied SL327 abolished forebrain pERK immunoreactivity, and significantly reduced cell death and associated microglial activation at 48 h post-HI. We then explored the effects of cell-specific ERK2 deletion alone or in combination with global ERK1 knockout under the same conditions of HI insult. Neuronal ERK2 deletion strongly decreased infarct size, neuronal cell death and microglial activation in grey matter following both HI alone or LPS-sensitised HI. ERK1 deletion attenuated the protective effect of neuronal ERK2 deletion. Removal of astroglial ERK2 produced a reverse response, with a 3- to 4-fold increase in microglial activation and cell death. Our data suggest a cell-specific and time-dependent role of ERK in neonatal HI, with a predominant, neurotoxic effect of neuronal ERK2, which is counteracted by neuroprotection by ERK1 and astrocytic ERK2. Overall, global pharmacological inhibition of ERK phosphorylation is strongly neuroprotective.


Subject(s)
Astrocytes/metabolism , Hypoxia-Ischemia, Brain/metabolism , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Neurons/metabolism , Animals , Animals, Newborn , Brain/metabolism , Brain/ultrastructure , Mice, Inbred C57BL , Mice, Transgenic , Mitogen-Activated Protein Kinase 1/genetics , Phosphorylation
4.
J Neurochem ; 136(5): 981-94, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26669927

ABSTRACT

Hypoxic-ischaemic encephalopathy is a leading cause of child death, with high mortality and morbidity, including cerebral palsy, epilepsy and cognitive disabilities. Hypoxia-ischaemia (HI) strongly up-regulates Signal Transducer and Activator of Transcription 3 (STAT3) in the immature brain. Our aim was to establish whether STAT3 up-regulation is associated with neonatal HI-brain damage and evaluate the phosphorylated STAT3-contribution from different cell types in eliciting damage. We subjected postnatal day seven mice to unilateral carotid artery ligation followed by 60 min hypoxia. Neuronal STAT3-deletion reduced cell death, tissue loss, microglial and astroglial activation in all brain regions. Astroglia-specific STAT3-deletion also reduced cell death, tissue loss and microglial activation, although not as strongly as the deletion in neurons. Systemic pre-insult STAT3-blockade at tyrosine 705 (Y705) with JAK2-inhibitor WP1066 reduced microglial and astroglial activation to a more moderate degree, but in a pattern similar to the one produced by the cell-specific deletions. Our results suggest that STAT3 is a crucial factor in neonatal HI-brain damage and its removal in neurons or astrocytes, and, to some extent, inhibition of its phosphorylation via JAK2-blockade reduces inflammation and tissue loss. Overall, the protective effects of STAT3 inactivation make it a possible target for a therapeutic strategy in neonatal HI. Current data show that neuronal and astroglial STAT3 molecules are involved in the pathways underlying cell death, tissue loss and gliosis following neonatal hypoxia-ischaemia, but differ with respect to the target of their effect. Y705-phosphorylation contributes to hypoxic-ischaemic histopathology. Protective effects of STAT3 inactivation make it a possible target for a therapeutic strategy in neonatal hypoxia-ischaemia.


Subject(s)
Brain/metabolism , Hypoxia-Ischemia, Brain/metabolism , Hypoxia/metabolism , Neurons/metabolism , STAT3 Transcription Factor/metabolism , Animals , Animals, Newborn , Cell Death/drug effects , Mice , Molecular Sequence Data , Signal Transduction/physiology , Up-Regulation
5.
Curr Pediatr Rev ; 10(1): 37-47, 2014.
Article in English | MEDLINE | ID: mdl-25055862

ABSTRACT

Neonatal brain injury remains a devastating condition, with poor outcomes despite the institution of an effective neuroprotective strategy of therapeutic hypothermia. There is an urgent need to develop additional neuroprotective strategies and to tailor our clinical predictive ability for families and their infants. Such goals could be more readily achieved if reliable early clinical indicators or biomarkers existed. This review will explore the relation between magnetic resonance (MR) imaging biomarkers and the degree of brain pathology observed in our translational piglet model of perinatal asphyxia. We also suggest biomarker relevance at a cellular level. The review will describe the development needed to optimize and simplify the use of biomarkers to speed up future trials of neuroprotection.


Subject(s)
Asphyxia Neonatorum/pathology , Hypothermia, Induced/methods , Hypoxia-Ischemia, Brain/pathology , Magnetic Resonance Spectroscopy , Neuroprotective Agents/pharmacology , Animals , Animals, Newborn , Aspartic Acid/analogs & derivatives , Aspartic Acid/metabolism , Asphyxia Neonatorum/metabolism , Biomarkers/metabolism , Choline/metabolism , Creatine/metabolism , Disease Models, Animal , Female , Humans , Hypoxia-Ischemia, Brain/metabolism , Infant, Newborn , Lactic Acid/metabolism , Pregnancy , Sensitivity and Specificity , Swine
6.
J Neurochem ; 130(4): 555-62, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24762056

ABSTRACT

Neonatal hypoxic ischaemic (HI) injury frequently causes neural impairment in surviving infants. Our knowledge of the underlying molecular mechanisms is still limited. Protein deimination is a post-translational modification caused by Ca(+2) -regulated peptidylarginine deiminases (PADs), a group of five isozymes that display tissue-specific expression and different preference for target proteins. Protein deimination results in altered protein conformation and function of target proteins, and is associated with neurodegenerative diseases, gene regulation and autoimmunity. In this study, we used the neonatal HI and HI/infection [lipopolysaccharide (LPS) stimulation] murine models to investigate changes in protein deimination. Brains showed increases in deiminated proteins, cell death, activated microglia and neuronal loss in affected brain areas at 48 h after hypoxic ischaemic insult. Upon treatment with the pan-PAD inhibitor Cl-amidine, a significant reduction was seen in microglial activation, cell death and infarct size compared with control saline or LPS-treated animals. Deimination of histone 3, a target protein of the PAD4 isozyme, was increased in hippocampus and cortex specifically upon LPS stimulation and markedly reduced following Cl-amidine treatment. Here, we demonstrate a novel role for PAD enzymes in neural impairment in neonatal HI Encephalopathy, highlighting their role as promising new candidates for drug-directed intervention in neurotrauma. Hypoxic Ischaemic Insult (HI) results in activation of peptidylarginine deiminases (PADs) because of calcium dysregulation. Target proteins undergo irreversible changes of protein bound arginine to citrulline, resulting in protein misfolding. Infection in synergy with HI causes up-regulation of TNFα, nuclear translocation of PAD4 and change in gene regulation as a result of histone deimination. Pharmacological PAD inhibition significantly reduced HI brain damage.


Subject(s)
Enzyme Inhibitors/pharmacology , Hydrolases/antagonists & inhibitors , Hypoxia-Ischemia, Brain/pathology , Hypoxia-Ischemia, Brain/prevention & control , Animals , Animals, Newborn , Brain Infarction/drug therapy , Brain Infarction/pathology , Cell Death/drug effects , Central Nervous System Bacterial Infections/drug therapy , Central Nervous System Bacterial Infections/pathology , Immunohistochemistry , In Situ Nick-End Labeling , Isoenzymes/antagonists & inhibitors , Isoenzymes/metabolism , Lipopolysaccharides , Mice , Mice, Inbred C57BL , Microglia/drug effects , Neurons/drug effects , Neurons/pathology , Ornithine/analogs & derivatives , Ornithine/toxicity , Protein-Arginine Deiminases
7.
Adv Exp Med Biol ; 812: 165-171, 2014.
Article in English | MEDLINE | ID: mdl-24729229

ABSTRACT

The relationship between cerebral autoregulation (CA) and the neurotoxic effects of anaesthesia with and without surgery is investigated. Newborn piglets were randomly assigned to receive either 6 h of anaesthesia (isoflurane) or the same with an additional hour of minor surgery. The effect of the spontaneous changes in mean arterial blood pressure (MABP) on the cerebral haemodynamics (oxy- and deoxy-haemoglobin, HbO2 and Hb) was measured using transverse broadband near-infrared spectroscopy (NIRS). A marker for impaired CA, concordance between MABP and intravascular oxygenation (HbD = HbO2 - Hb) in the ultra-low frequency domain (0.0018-0.0083 Hz), was assessed using coherence analysis. Presence of CA impairment was not significant but found to increase with surgical exacerbation. The impairment did not correlate with histological outcome (presence of cell death, apoptosis and microglial activation in the brain).


Subject(s)
Anesthesia , Brain/physiology , Surgical Procedures, Operative , Animals , Animals, Newborn , Brain/blood supply , Spectroscopy, Near-Infrared , Swine
8.
Brain ; 136(Pt 1): 90-105, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23183236

ABSTRACT

Despite treatment with therapeutic hypothermia, almost 50% of infants with neonatal encephalopathy still have adverse outcomes. Additional treatments are required to maximize neuroprotection. Melatonin is a naturally occurring hormone involved in physiological processes that also has neuroprotective actions against hypoxic-ischaemic brain injury in animal models. The objective of this study was to assess neuroprotective effects of combining melatonin with therapeutic hypothermia after transient hypoxia-ischaemia in a piglet model of perinatal asphyxia using clinically relevant magnetic resonance spectroscopy biomarkers supported by immunohistochemistry. After a quantified global hypoxic-ischaemic insult, 17 newborn piglets were randomized to the following: (i) therapeutic hypothermia (33.5°C from 2 to 26 h after resuscitation, n = 8) and (ii) therapeutic hypothermia plus intravenous melatonin (5 mg/kg/h over 6 h started at 10 min after resuscitation and repeated at 24 h, n = 9). Cortical white matter and deep grey matter voxel proton and whole brain (31)P magnetic resonance spectroscopy were acquired before and during hypoxia-ischaemia, at 24 and 48 h after resuscitation. There was no difference in baseline variables, insult severity or any physiological or biochemical measure, including mean arterial blood pressure and inotrope use during the 48 h after hypoxia-ischaemia. Plasma levels of melatonin were 10 000 times higher in the hypothermia plus melatonin than hypothermia alone group. Melatonin-augmented hypothermia significantly reduced the hypoxic-ischaemic-induced increase in the area under the curve for proton magnetic resonance spectroscopy lactate/N-acetyl aspartate and lactate/total creatine ratios in the deep grey matter. Melatonin-augmented hypothermia increased levels of whole brain (31)P magnetic resonance spectroscopy nucleotide triphosphate/exchangeable phosphate pool. Correlating with improved cerebral energy metabolism, TUNEL-positive nuclei were reduced in the hypothermia plus melatonin group compared with hypothermia alone in the thalamus, internal capsule, putamen and caudate, and there was reduced cleaved caspase 3 in the thalamus. Although total numbers of microglia were not decreased in grey or white matter, expression of the prototypical cytotoxic microglial activation marker CD86 was decreased in the cortex at 48 h after hypoxia-ischaemia. The safety and improved neuroprotection with a combination of melatonin with cooling support phase II clinical trials in infants with moderate and severe neonatal encephalopathy.


Subject(s)
Brain/drug effects , Hypothermia, Induced/methods , Hypoxia-Ischemia, Brain/therapy , Melatonin/therapeutic use , Neuroprotective Agents/therapeutic use , Animals , Animals, Newborn , Asphyxia Neonatorum/metabolism , Asphyxia Neonatorum/pathology , Asphyxia Neonatorum/therapy , Blood Pressure/drug effects , Blood Pressure/physiology , Brain/metabolism , Brain/pathology , Disease Models, Animal , Energy Metabolism/drug effects , Energy Metabolism/physiology , Humans , Hypoxia-Ischemia, Brain/metabolism , Hypoxia-Ischemia, Brain/pathology , Infant, Newborn , Magnetic Resonance Spectroscopy , Male , Melatonin/blood , Melatonin/pharmacology , Neuroprotective Agents/pharmacology , Resuscitation , Swine , Treatment Outcome
9.
J Neurochem ; 124(5): 645-57, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23171224

ABSTRACT

Na⁺/H⁺ exchanger (NHE) blockade attenuates the detrimental consequences of ischaemia and reperfusion in myocardium and brain in adult and neonatal animal studies. Our aim was to use magnetic resonance spectroscopy (MRS) biomarkers and immunohistochemistry to investigate the cerebral effects of the NHE inhibitor, methyl isobutyl amiloride (MIA) given after severe perinatal asphyxia in the piglet. Eighteen male piglets (aged < 24 h) underwent transient global cerebral hypoxia-ischaemia and were randomized to (i) saline placebo; or (ii) 3 mg/kg intravenous MIA administered 10 min post-insult and 8 hourly thereafter. Serial phosphorus-31 (³¹P) and proton (¹H) MRS data were acquired before, during and up to 48 h after hypoxia-ischaemia and metabolite-ratio time-series Area under the Curve (AUC) calculated. At 48 h, histological and immunohistochemical assessments quantified regional tissue injury. MIA decreased thalamic lactate/N-acetylaspartate and lactate/creatine AUCs (both p < 0.05) compared with placebo. Correlating with improved cerebral energy metabolism, transferase mediated biotinylated d-UTP nick end-labelling (TUNEL) positive cell density was reduced in the MIA group in cerebral cortex, thalamus and white matter (all p < 0.05) and caspase 3 immunoreactive cells were reduced in pyriform cortex and caudate nucleus (both p < 0.05). Microglial activation was reduced in pyriform and midtemporal cortex (both p < 0.05). Treatment with MIA starting 10 min after hypoxia-ischaemia was neuroprotective in this perinatal asphyxia model.


Subject(s)
Amiloride/analogs & derivatives , Asphyxia/drug therapy , Hypoxia-Ischemia, Brain/drug therapy , Microglia/drug effects , Neuroprotective Agents/pharmacology , Amiloride/pharmacology , Animals , Aspartic Acid/analogs & derivatives , Aspartic Acid/metabolism , Asphyxia/metabolism , Brain/drug effects , Brain/metabolism , Cell Death/drug effects , Disease Models, Animal , Hypoxia-Ischemia, Brain/metabolism , Immunohistochemistry , In Situ Nick-End Labeling , Lactic Acid/metabolism , Magnetic Resonance Spectroscopy , Male , Microglia/metabolism , Swine
10.
Neuron ; 75(4): 633-47, 2012 Aug 23.
Article in English | MEDLINE | ID: mdl-22920255

ABSTRACT

The radical response of peripheral nerves to injury (Wallerian degeneration) is the cornerstone of nerve repair. We show that activation of the transcription factor c-Jun in Schwann cells is a global regulator of Wallerian degeneration. c-Jun governs major aspects of the injury response, determines the expression of trophic factors, adhesion molecules, the formation of regeneration tracks and myelin clearance and controls the distinctive regenerative potential of peripheral nerves. A key function of c-Jun is the activation of a repair program in Schwann cells and the creation of a cell specialized to support regeneration. We show that absence of c-Jun results in the formation of a dysfunctional repair cell, striking failure of functional recovery, and neuronal death. We conclude that a single glial transcription factor is essential for restoration of damaged nerves, acting to control the transdifferentiation of myelin and Remak Schwann cells to dedicated repair cells in damaged tissue.


Subject(s)
Nerve Regeneration/physiology , Proto-Oncogene Proteins c-jun/metabolism , Schwann Cells/metabolism , Sciatic Neuropathy/pathology , Adenoviridae/genetics , Analysis of Variance , Animals , Benzofurans , Cell Movement/genetics , Disease Models, Animal , Gene Expression Regulation/genetics , Genetic Vectors/physiology , Macrophages/metabolism , Macrophages/pathology , Macrophages/ultrastructure , Mice , Mice, Transgenic , Microfluidic Analytical Techniques , Microscopy, Electron, Transmission , Motor Neurons/metabolism , Motor Neurons/pathology , Motor Neurons/ultrastructure , Myelin Sheath/pathology , Myelin Sheath/ultrastructure , Proto-Oncogene Proteins c-jun/genetics , Schwann Cells/pathology , Schwann Cells/ultrastructure , Sciatic Neuropathy/metabolism , Sciatic Neuropathy/physiopathology , Sciatic Neuropathy/therapy , Spinal Cord/pathology
11.
J Cell Biol ; 198(1): 127-41, 2012 Jul 09.
Article in English | MEDLINE | ID: mdl-22753894

ABSTRACT

The AP-1 transcription factor c-Jun is a master regulator of the axonal response in neurons. c-Jun also functions as a negative regulator of myelination in Schwann cells (SCs) and is strongly reactivated in SCs upon axonal injury. We demonstrate here that, after injury, the absence of c-Jun specifically in SCs caused impaired axonal regeneration and severely increased neuronal cell death. c-Jun deficiency resulted in decreased expression of several neurotrophic factors, and GDNF and Artemin, both of which encode ligands for the Ret receptor tyrosine kinase, were identified as novel direct c-Jun target genes. Genetic inactivation of Ret specifically in neurons resulted in regeneration defects without affecting motoneuron survival and, conversely, administration of recombinant GDNF and Artemin protein substantially ameliorated impaired regeneration caused by c-Jun deficiency. These results reveal an unexpected function for c-Jun in SCs in response to axonal injury, and identify paracrine Ret signaling as an important mediator of c-Jun function in SCs during regeneration.


Subject(s)
Axons/physiology , Motor Neurons/physiology , Nerve Regeneration/physiology , Paracrine Communication/physiology , Proto-Oncogene Proteins c-jun/physiology , Schwann Cells/physiology , Animals , Cell Survival , Down-Regulation/physiology , Glial Cell Line-Derived Neurotrophic Factor/pharmacology , Mice , Nerve Tissue Proteins/physiology
12.
Cell Tissue Res ; 349(1): 15-26, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22580509

ABSTRACT

The robust axon regeneration that occurs following peripheral nerve injury is driven by transcriptional activation of the regeneration program and by the expression of a wide range of downstream effector molecules from neuropeptides and neurotrophic factors to adhesion molecules and cytoskeletal adaptor proteins. These regeneration-associated effector molecules regulate the actin-tubulin machinery of growth-cones, integrate intracellular signalling and stimulatory and inhibitory signals from the local environment and translate them into axon elongation. In addition to the neuronally derived molecules, an important transcriptional component is found in locally activated Schwann cells and macrophages, which release a number of cytokines, growth factors and neurotrophins that support neuronal survival and axonal regeneration and that might provide directional guidance cues towards appropriate peripheral targets. This review aims to provide a comprehensive up-to-date account of the transcriptional regulation and functional role of these effector molecules and of the information that they can give us with regard to the organisation of the regeneration program.


Subject(s)
Nerve Regeneration/physiology , Peripheral Nerves/metabolism , Peripheral Nerves/physiology , Signal Transduction , Animals , Chromatin/metabolism , Growth Cones/metabolism , Humans , Nerve Regeneration/genetics , Signal Transduction/genetics , Transcription, Genetic
13.
J Neurochem ; 121(4): 607-18, 2012 May.
Article in English | MEDLINE | ID: mdl-22372722

ABSTRACT

Although neural c-Jun is essential for successful peripheral nerve regeneration, the cellular basis of this effect and the impact of c-Jun activation are incompletely understood. In the current study, we explored the effects of neuron-selective c-Jun deletion, substitution of serine 63 and 73 phosphoacceptor sites with non-phosphorylatable alanine, and deletion of Jun N-terminal kinases 1, 2 and 3 in mouse facial nerve regeneration. Removal of the floxed c-jun gene in facial motoneurons using cre recombinase under control of a neuron-specific synapsin promoter (junΔS) abolished basal and injury-induced neuronal c-Jun immunoreactivity, as well as most of the molecular responses following facial axotomy. Absence of neuronal Jun reduced the speed of axonal regeneration following crush, and prevented most cut axons from reconnecting to their target, significantly reducing functional recovery. Despite blocking cell death, this was associated with a large number of shrunken neurons. Finally, junΔS mutants also had diminished astrocyte and microglial activation and T-cell influx, suggesting that these non-neuronal responses depend on the release of Jun-dependent signals from neighboring injured motoneurons. The effects of substituting serine 63 and 73 phosphoacceptor sites (junAA), or of global deletion of individual kinases responsible for N-terminal c-Jun phosphorylation were mild. junAA mutants showed decrease in neuronal cell size, a moderate reduction in post-axotomy CD44 levels and slightly increased astrogliosis. Deletion of Jun N-terminal kinase (JNK)1 or JNK3 showed delayed functional recovery; deletion of JNK3 also interfered with T-cell influx, and reduced CD44 levels. Deletion of JNK2 had no effect. Thus, neuronal c-Jun is needed in regeneration, but JNK phosphorylation of the N-terminus mostly appears to not be required for its function.


Subject(s)
Axons/physiology , Nerve Regeneration/physiology , Neurons/physiology , Proto-Oncogene Proteins c-jun/physiology , Animals , Atrophy , Axons/ultrastructure , Cell Death , Female , Hyaluronan Receptors/metabolism , Male , Mice , Mice, Knockout , Mitogen-Activated Protein Kinase 10/genetics , Mitogen-Activated Protein Kinase 10/physiology , Mitogen-Activated Protein Kinase 8/genetics , Mitogen-Activated Protein Kinase 8/physiology , Mitogen-Activated Protein Kinase 9/genetics , Mitogen-Activated Protein Kinase 9/physiology , Motor Neurons/physiology , Nerve Regeneration/genetics , Neurons/ultrastructure , Phosphorylation , Point Mutation/physiology , Proto-Oncogene Proteins c-jun/genetics
14.
Front Mol Neurosci ; 5: 8, 2012.
Article in English | MEDLINE | ID: mdl-22363260

ABSTRACT

Following axotomy, the activation of multiple intracellular signaling cascades causes the expression of a cocktail of regeneration-associated transcription factors which interact with each other to determine the fate of the injured neurons. The nerve injury response is channeled through manifold and parallel pathways, integrating diverse inputs, and controlling a complex transcriptional output. Transcription factors form a vital link in the chain of regeneration, converting injury-induced stress signals into downstream protein expression via gene regulation. They can regulate the intrinsic ability of axons to grow, by controlling expression of whole cassettes of gene targets. In this review, we have investigated the functional roles of a number of different transcription factors - c-Jun, activating transcription factor 3, cAMP response element binding protein, signal transducer, and activator of transcription-3, CCAAT/enhancer binding proteins ß and δ, Oct-6, Sox11, p53, nuclear factor kappa-light-chain-enhancer of activated B cell, and ELK3 - in peripheral nerve regeneration. Studies involving use of conditional mutants, microarrays, promoter region mapping, and different injury paradigms, have enabled us to understand their distinct as well as overlapping roles in achieving anatomical and functional regeneration after peripheral nerve injury.

15.
Front Mol Neurosci ; 5: 7, 2012.
Article in English | MEDLINE | ID: mdl-22347845

ABSTRACT

Activating transcription factor 3 (ATF3) belongs to the ATF/cyclic AMP responsive element binding family of transcription factors and is often described as an adaptive response gene whose activity is usually regulated by stressful stimuli. Although expressed in a number of splice variants and generally recognized as a transcriptional repressor, ATF3 has the ability to interact with a number of other transcription factors including c-Jun to form complexes which not only repress, but can also activate various genes. ATF3 expression is modulated mainly at the transcriptional level and has markedly different effects in different types of cell. The levels of ATF3 mRNA and protein are normally very low in neurons and glia but their expression is rapidly upregulated in response to injury. ATF3 expression in neurons is closely linked to their survival and the regeneration of their axons following axotomy, and that in peripheral nerves correlates with the generation of a Schwann cell phenotype that is conducive to axonal regeneration. ATF3 is also induced by Toll-like receptor (TLR) ligands but acts as a negative regulator of TLR signaling, suppressing the innate immune response which is involved in immuno-surveillance and can enhance or reduce the survival of injured neurons and promote the regeneration of their axons.

16.
Ann Neurol ; 70(1): 133-50, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21674582

ABSTRACT

OBJECTIVE: Additional treatments for therapeutic hypothermia are required to maximize neuroprotection for perinatal asphyxial encephalopathy. We assessed neuroprotective effects of combining inhaled xenon with therapeutic hypothermia after transient cerebral hypoxia-ischemia in a piglet model of perinatal asphyxia using magnetic resonance spectroscopy (MRS) biomarkers supported by immunohistochemistry. METHODS: Thirty-six newborn piglets were randomized (all groups n = 9), with intervention from 2 to 26 hours, to: (1) normothermia; (2) normothermia + 24 hours 50% inhaled xenon; (3) 24 hours hypothermia (33.5°C); or (4) 24 hours hypothermia (33.5°C) + 24 hours 50% inhaled xenon. Serial MRS was acquired before, during, and up to 48 hours after hypoxia-ischemia. RESULTS: Mean arterial blood pressure was lower in all treatment groups compared with normothermia (p < 0.01) (although >40mmHg); the combined therapy group required more fluid boluses (p < 0.05) and inotropes (p < 0.001). Compared with no intervention, both hypothermia and xenon-augmented hypothermia reduced the temporal regression slope magnitudes for phosphorus-MRS inorganic phosphate/exchangeable phosphate pool (EPP) and phosphocreatine/EPP (both p < 0.05); for lactate/N-acetylaspartate (NAA), only xenon-augmented hypothermia reduced the slope (p < 0.01). Xenon-augmented hypothermia also reduced transferase-mediated deoxyuridine triphosphate nick-end labeling (TUNEL)(+) nuclei and caspase 3 immunoreactive cells in parasagittal cortex and putamen and increased microglial ramification in midtemporal cortex compared with the no treatment group (p < 0.05). Compared with hypothermia, however, combination treatment did not reach statistical significance for any measure. Lactate/NAA showed a strong positive correlation with TUNEL; nucleotide triphosphate/EPP showed a strong negative correlation with microglial ramification (both p < 0.01). INTERPRETATION: Compared with no treatment, xenon-augmented hypothermia reduced cerebral MRS abnormalities and cell death markers in some brain regions. Compared with hypothermia, xenon-augmented hypothermia did not reach statistical significance for any measure. The safety and possible improved efficacy support phase II trials.


Subject(s)
Aspartic Acid/analogs & derivatives , Asphyxia/metabolism , Asphyxia/therapy , Hypothermia, Induced/methods , Lactic Acid/metabolism , Xenon/administration & dosage , Administration, Inhalation , Animals , Animals, Newborn , Aspartic Acid/antagonists & inhibitors , Aspartic Acid/metabolism , Cell Death/drug effects , Cell Death/physiology , Lactic Acid/antagonists & inhibitors , Male , Random Allocation , Swine , Time Factors
17.
Ann Anat ; 193(4): 248-58, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21501955

ABSTRACT

The peripheral nervous system is known to regenerate comparatively well and this ability is mirrored in the de novo expression or upregulation of a wide variety of molecules involved in axonal outgrowth starting with transcription factors, but also including growth-stimulating substances, guidance and cell adhesion molecules, intracellular signaling enzymes and proteins involved in regulating cell-surface cytoskeletal interactions. Recent studies using pharmacological agents, and global as well as neuron-selective gene inactivation techniques have shed light on those endogenous molecules that play a non-redundant role in mediating regenerative axonal outgrowth in vivo. The aim of the current review is to sketch the sequence of molecular events from early sensors of injury to transcription factors to downstream effectors that cooperate in successful regeneration and functional recovery.


Subject(s)
Nerve Regeneration/physiology , Peripheral Nerves/physiology , Recovery of Function/physiology , Signal Transduction/physiology , Transcription Factors/physiology , Axons/physiology , Humans , Intracellular Signaling Peptides and Proteins/physiology , Peripheral Nervous System/physiopathology
18.
Dev Neurosci ; 33(6): 505-18, 2011.
Article in English | MEDLINE | ID: mdl-22343485

ABSTRACT

We assessed the distribution in brain pH after neonatal hypoxic-ischaemic insult and its correlation with local injury. Postnatal day 7 mice were injected with neutral red and underwent left carotid occlusion and exposure to 8% oxygen. Images captured from the cut surface of snap-frozen brain were used to calculate the pH from the blue-green absorbance ratios. Carotid occlusion alone had no effect, but combined with hypoxia caused rapid, biphasic pH decline, with the first plateau at 15-30 min, and the second at 60-90 min. The ipsilateral dorsal cortex, hippocampus, striatum and thalamus were most affected. Contralateral pH initially showed only 30% of the ipsilateral decline, becoming more acidotic with increasing duration. Systemic blood analysis revealed, compared with hypoxia alone, that combined insult caused a 63% decrease in blood glucose (1.3 ± 0.2 mM), a 2-fold increase in circulating lactate (17.7 ± 2.9 mM), a reduction in CO(2) to 1.9 ± 0.1 kPa and a drop in pH (7.26 ± 0.06). Re-oxygenation resulted in the normalisation of systemic changes, as well as a global alkaline rebound in brain pH at 4-6 h. A topographic comparison of brain injury showed only a partial correlation with pH changes, with the severest injury occurring in the ipsilateral hippocampus and sparing acidic parts of the contralateral cortex.


Subject(s)
Brain/physiopathology , Hypoxia-Ischemia, Brain/blood , Hypoxia-Ischemia, Brain/physiopathology , Animals , Animals, Newborn , Female , Functional Laterality , Hydrogen-Ion Concentration , Immunohistochemistry , Mice , Mice, Inbred C57BL
19.
Lab Invest ; 91(3): 328-41, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21135813

ABSTRACT

In the current study, we explored the role of TNF cluster cytokines on the lipopolysaccharide (LPS)-mediated, synergistic increase in brain injury after hypoxic ischemic insult in postnatal day 7 mice. Pretreatment with moderate doses of LPS (0.3 µg/g) resulted in particularly pronounced synergistic injury within 12 h. Systemic application of LPS alone resulted in a strong upregulation of inflammation-associated cytokines TNFα, LTß, interleukin (IL) 1ß, IL6, chemokines, such as CXCL1, and adhesion molecules E-Selectin, P-Selectin and intercellular adhesion molecule-1 (ICAM1), as well as a trend toward increased LTα levels in day 7 mouse forebrain. In addition, it was also associated with strong activation of brain blood vessel endothelia and local microglial cells. Here, deletion of the entire TNF gene cluster, removing TNFα, LTß and LTα completely abolished endotoxin-mediated increase in the volume of cerebral infarct. Interestingly, the same deletion also prevented endothelial and microglial activation following application of LPS alone, suggesting the involvement of these cell types in bringing about the LPS-mediated sensitization to neonatal brain injury.


Subject(s)
Brain/metabolism , Disease Susceptibility , Hypoxia-Ischemia, Brain/metabolism , Lipopolysaccharides/toxicity , Lymphotoxin-alpha/metabolism , Lymphotoxin-beta/metabolism , Tumor Necrosis Factor-alpha/metabolism , Animals , Animals, Newborn , Brain/growth & development , Brain/pathology , Cell Adhesion Molecules/genetics , Cell Adhesion Molecules/metabolism , Cerebral Infarction/chemically induced , Cerebral Infarction/pathology , Cytokines/genetics , Cytokines/metabolism , Endothelium, Vascular/growth & development , Endothelium, Vascular/metabolism , Gene Expression Regulation, Developmental , Hypoxia-Ischemia, Brain/mortality , Hypoxia-Ischemia, Brain/pathology , Lymphotoxin-alpha/genetics , Lymphotoxin-beta/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Microglia/metabolism , Multigene Family , RNA, Messenger/metabolism , Sequence Deletion , Severity of Illness Index , Survival Analysis , Tumor Necrosis Factor-alpha/genetics
20.
J Biol Chem ; 285(49): 38534-42, 2010 Dec 03.
Article in English | MEDLINE | ID: mdl-20889973

ABSTRACT

Progressive accumulation of DNA damage is causally involved in cellular senescence and organismal aging. The DNA damage kinase ATM plays a central role in maintaining genomic stability. ATM mutations cause the genetic disorder ataxia telangiectasia, which is primarily characterized by progressive neurodegeneration and cancer susceptibility. Although the importance of ATM function to protect against oxidative DNA damage and during aging is well described, the mechanism of ATM activation by these stimuli is not known. Here we identify ATM interactor (ATMIN) as an essential component of the ATM signaling pathway in response to oxidative stress and aging. Embryos lacking ATMIN (atmin(Δ/Δ)) died in utero and showed increased numbers of cells positive for phosphorylated histone H2aX, indicative of increased DNA damage. atmin(Δ/Δ) mouse embryonic fibroblasts accumulated DNA damage and prematurely entered senescence when cultured at atmospheric oxygen levels (20%), but this defect was rescued by addition of an antioxidant and also by culturing cells at physiological oxygen levels (3%). In response to acute oxidative stress, atmin(Δ/Δ) mouse embryonic fibroblasts showed slightly lower levels of ATM phosphorylation and reduced ATM substrate phosphorylation. Conditional deletion of ATMIN in the murine nervous system (atmin(ΔN)) resulted in reduced numbers of dopaminergic neurons, as does ATM deficiency. ATM activity was observed in old, but not in young, control mice, but aging-induced ATM signaling was impaired by ATMIN deficiency. Consequently, old atmin(ΔN) mice showed accumulation of DNA damage in the cortex accompanied by gliosis, resulting in increased mortality of aging mutant mice. These results suggest that ATMIN mediates ATM activation by oxidative stress, and thereby ATMIN protects the aging brain by preventing accumulation of DNA damage.


Subject(s)
Aging/metabolism , Carrier Proteins/metabolism , Cell Cycle Proteins/metabolism , Cerebral Cortex/metabolism , DNA Damage/physiology , DNA-Binding Proteins/metabolism , Nuclear Proteins/metabolism , Oxidative Stress/physiology , Protein Serine-Threonine Kinases/metabolism , Tumor Suppressor Proteins/metabolism , Aging/genetics , Aging/pathology , Animals , Antioxidants/metabolism , Ataxia Telangiectasia Mutated Proteins , Carrier Proteins/genetics , Cell Cycle Proteins/genetics , Cells, Cultured , Cellular Senescence/genetics , Cerebral Cortex/embryology , Cerebral Cortex/pathology , DNA-Binding Proteins/genetics , Embryo Loss/genetics , Embryo Loss/metabolism , Embryo Loss/pathology , Embryo, Mammalian/metabolism , Embryo, Mammalian/pathology , Fibroblasts/metabolism , Fibroblasts/pathology , Histones/genetics , Histones/metabolism , Mice , Mice, Mutant Strains , Neurons/metabolism , Neurons/pathology , Nuclear Proteins/genetics , Oxygen/metabolism , Phosphorylation/genetics , Protein Serine-Threonine Kinases/genetics , Transcription Factors , Tumor Suppressor Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...