Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 32
Filter
1.
Health Promot Int ; 39(4)2024 Aug 01.
Article in English | MEDLINE | ID: mdl-38980690

ABSTRACT

Accessible, up-to-date information on traumatic brain injury (TBI) can be challenging to find and is needed to address TBI knowledge gaps and improve outcomes for people who experience a TBI. The Understanding TBI Massive Open Online Course (TBI MOOC) was developed to increase TBI knowledge across a diverse global audience. We sought to characterize the TBI MOOC participant cohort, to understand the reach of the course among this target audience. Examining the characteristics of TBI MOOC enrollees showed that participants came from a wide range of demographic backgrounds, had a variety of TBI experiences and had multiple reasons for enrolling in the MOOC. The majority of course participants shared some characteristics with other groups of health information seekers. Four distinct demographic profiles were identified among TBI MOOC participants (education seekers, TBI-aware participants, TBI care providers and retirees) using a novel approach combining chi-squared tests and network modularity. Participants assigned to the TBI-aware and retiree profiles were most likely to complete all modules of the MOOC, and the TBI-aware profile was more highly represented in more recent iterations of the MOOC. Together, these data indicate that the TBI MOOC provided information to a wide range of people, and particularly engaged participants with personal or family experience of TBI. However, engagement with this course was minimal among some hard-to-reach populations, including men and people with low levels of education, indicating that additional strategies are needed to ensure equity in health promotion.


Subject(s)
Brain Injuries, Traumatic , Humans , Brain Injuries, Traumatic/psychology , Male , Female , Adult , Middle Aged , Internet , Information Seeking Behavior , Health Knowledge, Attitudes, Practice , Aged
2.
J Neurotrauma ; 40(1-2): 4-21, 2023 01.
Article in English | MEDLINE | ID: mdl-35880422

ABSTRACT

The identification of effective pharmacotherapies for traumatic brain injury (TBI) remains a major challenge. Treatment with heparin and its derivatives is associated with neuroprotective effects after experimental TBI; however, the optimal dosage and method of administration, modes of action, and effects on hemorrhage remain unclear. Therefore, this review aimed to systematically evaluate, analyze, and summarize the available literature on the use of heparin and low molecular weight heparins (LMWHs) as treatment options for experimental TBI. We searched two online databases (PubMed and ISI Web of Science) to identify relevant studies. Data pertaining to TBI paradigm, animal subjects, drug administration, and all pathological and behavior outcomes were extracted. Eleven studies met our pre-specified inclusion criteria, and for outcomes with sufficient numbers, data from seven publications were analyzed in a weighted mean difference meta-analysis using a random-effects model. Study quality and risk of bias were also determined. Meta-analysis revealed that heparin and its derivatives decreased brain edema, leukocyte rolling, and vascular permeability, and improved neurological function. Further, treatment did not aggravate hemorrhage. These findings must be interpreted with caution, however, because they were determined from a limited number of studies with substantial heterogeneity. Also, overall study quality was low based on absences of data reporting, and potential publication bias was identified. Importantly, we found that there are insufficient data to evaluate the variables we had hoped to investigate. The beneficial effects of heparin and LMWHs, however, suggest that further pre-clinical studies are warranted.


Subject(s)
Brain Edema , Brain Injuries, Traumatic , Animals , Brain Edema/drug therapy , Brain Injuries, Traumatic/drug therapy , Hemorrhage/drug therapy , Heparin/toxicity , Heparin, Low-Molecular-Weight/pharmacology , Heparin, Low-Molecular-Weight/therapeutic use
3.
Int J Mol Sci ; 22(3)2021 Jan 20.
Article in English | MEDLINE | ID: mdl-33498409

ABSTRACT

Diabetic retinopathy (DR), one of the leading causes of blindness, is mainly diagnosed based on the vascular pathology of the disease. Current treatment options largely focus on this aspect with mostly insufficient therapeutic long-term efficacy. Mounting evidence implicates mitochondrial dysfunction and oxidative stress in the central etiology of DR. Consequently, drug candidates that aim at normalizing mitochondrial function could be an attractive therapeutic approach. This study compared the mitoprotective compounds, idebenone and elamipretide, side-by-side against two novel short-chain quinones (SCQs) in a rat model of DR. The model effectively mimicked type 2 diabetes over 21 weeks. During this period, visual acuity was monitored by measuring optokinetic response (OKR). Vision loss occurred 5-8 weeks after the onset of hyperglycemia. After 10 weeks of hyperglycemia, visual function was reduced by 65%. From this point, the right eyes of the animals were topically treated once daily with the test compounds. The left, untreated eye served as an internal control. Only three weeks of topical treatment significantly restored vision from 35% to 58-80%, while visual acuity of the non-treated eyes continued to deteriorate. Interestingly, the two novel SCQs restored visual acuity better than idebenone or elamipretide. This was also reflected by protection of retinal pathology against oxidative damage, retinal ganglion cell loss, reactive gliosis, vascular leakage, and retinal thinning. Overall, mitoprotective and, in particular, SCQ-based compounds have the potential to be developed into effective and fast-acting drug candidates against DR.


Subject(s)
Antioxidants/therapeutic use , Diabetic Retinopathy/drug therapy , Ubiquinone/analogs & derivatives , Animals , Antioxidants/pharmacology , Male , Mitochondria/drug effects , Oligopeptides/pharmacology , Oligopeptides/therapeutic use , Rats , Rats, Long-Evans , Ubiquinone/pharmacology , Ubiquinone/therapeutic use , Vision, Ocular
4.
J Neurosci Res ; 98(10): 1905-1932, 2020 10.
Article in English | MEDLINE | ID: mdl-32557778

ABSTRACT

In Alzheimer's disease, amyloid plaque formation is associated with the focal death of oligodendrocytes and soluble amyloid ß impairs the survival of oligodendrocytes in vitro. However, the response of oligodendrocyte progenitor cells (OPCs) to early amyloid pathology remains unclear. To explore this, we performed a histological, electrophysiological, and behavioral characterization of transgenic mice expressing a pathological form of human amyloid precursor protein (APP), containing three single point mutations associated with the development of familial Alzheimer's disease (PDGFB-APPSw.Ind , also known as J20 mice). PDGFB-APPSw.Ind transgenic mice had impaired survival from weaning, were hyperactive by 2 months of age, and developed amyloid plaques by 6 months of age, however, their spatial memory remained intact over this time course. Hippocampal OPC density was normal in P60-P180 PDGFB-APPSw.Ind transgenic mice and, by performing whole-cell patch-clamp electrophysiology, we found that their membrane properties, including their response to kainate (100 µM), were largely normal. However, by P100, the response of hippocampal OPCs to GABA was elevated in PDGFB-APPSw.Ind transgenic mice. We also found that the nodes of Ranvier were shorter, the paranodes longer, and the myelin thicker for hippocampal axons in young adult PDGFB-APPSw.Ind transgenic mice compared with wildtype littermates. Additionally, oligodendrogenesis was normal in young adulthood, but increased in the hippocampus, entorhinal cortex, and fimbria of PDGFB-APPSw.Ind transgenic mice as pathology developed. As the new oligodendrocytes were not associated with a change in total oligodendrocyte number, these cells are likely required for cell replacement.


Subject(s)
Amyloidosis/pathology , Brain/pathology , Myelin Sheath/pathology , Neurogenesis/physiology , Oligodendroglia/pathology , Age Factors , Amyloidosis/genetics , Animals , Female , Humans , Male , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Transgenic , Myelin Sheath/genetics
5.
J Neurotrauma ; 37(5): 782-791, 2020 03 01.
Article in English | MEDLINE | ID: mdl-32046575

ABSTRACT

Traumatic brain injury (TBI) can cause persistent cognitive changes and ongoing neurodegeneration in the brain. Accumulating epidemiological and pathological evidence implicates TBI in the development of Alzheimer's disease, the most common cause of dementia. Further, the TBI-induced form of dementia, called chronic traumatic encephalopathy, shares many pathological hallmarks present in multiple different diseases which cause dementia. The inflammatory and neuritic responses to TBI and dementia overlap, indicating that they may share common pathological mechanisms and that TBI may ultimately cause a pathological cascade culminating in the development of dementia. This review explores Australian pre-clinical research investigating the pathological links between TBI and dementia.


Subject(s)
Brain Injuries, Traumatic/pathology , Brain/pathology , Dementia/pathology , Animals , Australia , Brain/metabolism , Brain Injuries, Traumatic/complications , Brain Injuries, Traumatic/metabolism , Dementia/etiology , Dementia/metabolism , Humans , Microglia/metabolism , Microglia/pathology , Plaque, Amyloid/metabolism , Plaque, Amyloid/pathology , tau Proteins/metabolism
6.
Acta Neuropathol ; 137(5): 731-755, 2019 05.
Article in English | MEDLINE | ID: mdl-30535946

ABSTRACT

This review recounts the definitions and research evidence supporting the multifaceted roles of neuroinflammation in the injured brain following trauma. We summarise the literature fluctuating from the protective and detrimental properties that cytokines, leukocytes and glial cells play in the acute and chronic stages of TBI, including the intrinsic factors that influence cytokine responses and microglial functions relative to genetics, sex, and age. We elaborate on the pros and cons that cytokines, chemokines, and microglia play in brain repair, specifically neurogenesis, and how such conflicting roles may be harnessed therapeutically to sustain the survival of new neurons. With a brief review of the clinical and experimental findings demonstrating early and chronic inflammation impacts on outcomes, we focus on the clinical conditions that may be amplified by neuroinflammation, ranging from acute seizures to chronic epilepsy, neuroendocrine dysfunction, dementia, depression, post-traumatic stress disorder and chronic traumatic encephalopathy. Finally, we provide an overview of the therapeutic agents that have been tested to reduce inflammation-driven secondary pathological cascades and speculate the future promise of alternative drugs.


Subject(s)
Brain Injuries, Traumatic/physiopathology , Inflammation/physiopathology , Animals , Brain Injuries, Traumatic/epidemiology , Brain Injuries, Traumatic/therapy , Humans , Inflammation/epidemiology , Inflammation/therapy , Neuroimmunomodulation
7.
Crit Care Med ; 46(4): 554-561, 2018 04.
Article in English | MEDLINE | ID: mdl-29278529

ABSTRACT

OBJECTIVE: To determine profiles of serum ubiquitin carboxy-terminal hydrolase L1 and phosphorylated neurofilament heavy-chain, examine whether erythropoietin administration reduce their concentrations, and whether biomarkers discriminate between erythropoietin and placebo treatment groups. DESIGN: Single-center, prospective observational study. SETTING: A sub-study of the erythropoietin-traumatic brain injury clinical trial, conducted at the Alfred Hospital, Melbourne, Australia. PATIENTS: Forty-four patients with moderate-to-severe traumatic brain injury. INTERVENTIONS: Epoetin alfa 40,000 IU or 1 mL sodium chloride 0.9 as subcutaneous injection within 24 hours of traumatic brain injury. MEASUREMENTS AND MAIN RESULTS: Ubiquitin carboxy-terminal hydrolase L1, phosphorylated neurofilament heavy-chain, and erythropoietin concentrations were measured in serum by enzyme-linked immunosorbent assay from D0 (within 24 hr of injury, prior to erythropoietin/vehicle administration) to D5. Biomarker concentrations were compared between injury severities, diffuse versus focal traumatic brain injury and erythropoietin or placebo treatment groups. Ubiquitin carboxy-terminal hydrolase L1 peaked at 146.0 ng/mL on D0, significantly decreased to 84.30 ng/mL on D1, and declined thereafter. Phosphorylated neurofilament heavy-chain levels were lowest at D0 and peaked on D5 at 157.9 ng/mL. D0 ubiquitin carboxy-terminal hydrolase L1 concentrations were higher in diffuse traumatic brain injury. Peak phosphorylated neurofilament heavy-chain levels on D3 and D4 correlated with Glasgow Outcome Score-Extended, predicting poor outcome. Erythropoietin did not reduce concentrations of ubiquitin carboxy-terminal hydrolase L1 or phosphorylated neurofilament heavy-chain. CONCLUSIONS: Serum ubiquitin carboxy-terminal hydrolase L1 and phosphorylated neurofilament heavy-chain increase after traumatic brain injury reflecting early neuronal and progressive axonal injury. Consistent with lack of improved outcome in traumatic brain injury patients treated with erythropoietin, biomarker concentrations and profiles were not affected by erythropoietin. Pharmacokinetics of erythropoietin suggest that the dose given was possibly too low to exert neuroprotection.


Subject(s)
Brain Injuries, Traumatic/drug therapy , Epoetin Alfa/pharmacology , Epoetin Alfa/therapeutic use , Erythropoietin/blood , Neurofilament Proteins/blood , Ubiquitin Thiolesterase/drug effects , Adult , Australia , Biomarkers , Double-Blind Method , Enzyme-Linked Immunosorbent Assay , Epoetin Alfa/pharmacokinetics , Female , Glasgow Coma Scale , Humans , Male , Middle Aged , Prospective Studies , Ubiquitin Thiolesterase/blood
8.
PLoS One ; 12(11): e0188305, 2017.
Article in English | MEDLINE | ID: mdl-29176798

ABSTRACT

The atypical chemokine receptor ACKR2 promotes resolution of acute inflammation by operating as a scavenger receptor for inflammatory CC chemokines in several experimental models of inflammatory disorders, however its role in the brain remains unclear. Based on our previous reports of increased expression of inflammatory chemokines and their corresponding receptors following traumatic brain injury (TBI), we hypothesised that ACKR2 modulates neuroinflammation following brain trauma and that its deletion exacerbates cellular inflammation and chemokine production. We demonstrate increased CCL2 and ACKR2 mRNA expression in post-mortem human brain, whereby ACKR2 mRNA levels correlated with later times post-TBI. This data is consistent with the transient upregulation of ACKR2 observed in mouse brain after closed head injury (CHI). As compared to WT animals, ACKR2-/- mice showed a higher mortality rate after CHI, while the neurological outcome in surviving mice was similar. At day 1 post-injury, ACKR2-/- mice displayed aggravated lesion volume and no differences in CCL2 expression and macrophage recruitment relative to WT mice. Reciprocal regulation of ACKR2 and CCL2 expression was explored in cultured astrocytes, which are recognized as the major source of CCL2 and also express ACKR2. ACKR2 mRNA increased as early as 2 hours after an inflammatory challenge in WT astrocytes. As expected, CCL2 expression also dramatically increased at 4 hours in WT astrocytes but was significantly lower in ACKR2-/- astrocytes, possibly indicating a co-regulation of CCL2 and ACKR2 in these cells. Conversely, in vivo, CCL2 mRNA/protein levels were increased similarly in ACKR2-/- and WT brains at 4 and 12 hours after CHI, in line with the lack of differences in cerebral macrophage recruitment and neurological recovery. In conclusion, ACKR2 is induced after TBI and has a significant impact on mortality and lesion development acutely following CHI, while its role in chemokine expression, macrophage activation, brain pathology, and neurological recovery at later time-points is minor. Concordant to evidence in multiple sclerosis experimental models, our data corroborate a distinct role for ACKR2 in cerebral inflammatory processes compared to its reported functions in peripheral tissues.


Subject(s)
Brain Injuries, Traumatic/metabolism , Brain Injuries, Traumatic/mortality , Receptors, Chemokine/metabolism , Animals , Astrocytes/metabolism , Astrocytes/pathology , Bone and Bones/pathology , Brain/metabolism , Brain/pathology , Brain/physiopathology , Brain Injuries, Traumatic/genetics , Brain Injuries, Traumatic/physiopathology , Cells, Cultured , Chemokine CCL2/genetics , Chemokine CCL2/metabolism , Gene Deletion , Humans , Inflammation/pathology , Macrophages/metabolism , Macrophages/pathology , Male , Mice, Inbred C57BL , Mortality , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, Chemokine/genetics , Recovery of Function , Up-Regulation/genetics
9.
Mater Sci Eng C Mater Biol Appl ; 71: 584-593, 2017 Feb 01.
Article in English | MEDLINE | ID: mdl-27987748

ABSTRACT

Engineered nanoparticles with multiple complementary imaging modalities are of great benefit to the rapid treatment and diagnosis of disease in various organs. Herein, we report the formulation of cubosomes and hexosomes that carry multiple amphiphilic imaging contrast agents in their self-assembled lipid bilayers. This is the first report of the use of both near infrared fluorescent (NIRF) imaging and gadolinium lipid based magnetic resonance (MR) imaging modalities in cubosomes and hexosomes. High-throughput screening was used to rapidly optimize formulations with desirable nano-architectures and low in vitro cytotoxicity. The dual-modal imaging nanoparticles in vivo biodistribution and organ specific contrast enhancement were then studied. The NIRF in vivo imaging results indicated accumulation of both cubosomes and hexosomes in the liver and spleen of mice up to 20h post-injection. Remarkably, the biodistribution of the nanoparticle formulations was affected by the mesophase (i.e. cubic or hexagonal), a finding of significant importance for the future use of these compounds, with hexosomes showing higher accumulation in the spleen than the liver compared to cubosomes. Furthermore, in vivo MRI data of animals injected with either type of lyotropic liquid crystal nanoparticle displayed enhanced contrast in the liver and spleen.


Subject(s)
Contrast Media , Magnetic Resonance Imaging , Nanoparticles/chemistry , Optical Imaging , Animals , CHO Cells , Contrast Media/chemistry , Contrast Media/pharmacokinetics , Contrast Media/pharmacology , Cricetulus , Humans , Male , Mice , U937 Cells
10.
PLoS One ; 11(4): e0153418, 2016.
Article in English | MEDLINE | ID: mdl-27071013

ABSTRACT

Traumatic brain injury (TBI) is frequently characterized by neuronal, axonal and myelin loss, reactive gliosis and neuroinflammation, often associated with functional deficits. Endogenous repair mechanisms include production of new neurons from precursor cells, but usually the new neurons fail to integrate and survive more than a few weeks. This is in part mediated by the toxic and inflammatory environment present in the injured brain which activates precursor cells to proliferate and differentiate but limits survival of the newborn progeny. Therefore, an understanding of mechanisms that regulate production and survival of newborn neurons and the neuroinflammatory response after brain injury may lead to therapeutic options to improve outcomes. Suppressor of Cytokine Signaling 2 (SOCS2) promotes hippocampal neurogenesis and survival of newborn neurons in the adult brain and regulates anti-inflammatory responses in the periphery, suggesting it may be a useful candidate to improve outcomes of TBI. In this study the functional and cellular responses of SOCS2 over-expressing transgenic (SOCS2Tg) mice were compared to wildtype littermates following mild or moderately severe TBI. Unlike wildtype controls, SOCS2Tg mice showed functional improvement on a ladder test, with a smaller lesion volume at 7d post injury and increased numbers of proliferative CD11b+ microglia/macrophages at 35d post-injury in the mild injury paradigm. At 7d post-moderately severe injury there was an increase in the area covered by cells expressing an anti-inflammatory M2 phenotype marker (CD206+) but no difference in cells with a pro-inflammatory M1 phenotype marker (CD16/32+). No effect of SOCS2 overexpression was observed in production or survival of newborn neurons, even in the presence of the neuroprotective agent erythropoietin (EPO). Therefore, SOCS2 may improve outcome of TBI in mice by regulating aspects of the neuroinflammatory response, promoting a more anti-inflammatory environment, although this was not sufficient to enhance survival of newborn cortical neurons.


Subject(s)
Brain Injuries/pathology , Brain Injuries/physiopathology , Microglia/pathology , Suppressor of Cytokine Signaling Proteins/metabolism , Animals , Astrocytes/pathology , Brain Injuries/diagnosis , Brain Injuries/metabolism , Cell Proliferation , Dentate Gyrus/pathology , Erythropoietin/metabolism , Female , Humans , Macrophages/pathology , Male , Mice , Mice, Transgenic , Motor Activity , Neurogenesis , Prognosis , Signal Transduction , Suppressor of Cytokine Signaling Proteins/genetics , Time Factors
11.
Neurochem Int ; 96: 62-8, 2016 06.
Article in English | MEDLINE | ID: mdl-26930584

ABSTRACT

Autophagy is a homeostatic process for recycling proteins and organelles that is increasingly being proposed as a therapeutic target for acute and chronic neurodegenerative diseases, including stroke. Confirmation that autophagy is present in the human brain after stroke is imperative before prospective therapies can begin the translational process into clinical trials. Our current study using human post-mortem tissue observed an increase in staining in microtubule-associated protein 1 light chain 3 (LC3), sequestosome 1 (SQSTM1; also known as p62) and the increased appearance of autophagic vesicles after stroke. These data confirm that alterations in autophagy take place in the human brain after stroke and suggest that targeting autophagic processes after stroke may have clinical significance.


Subject(s)
Autophagy/physiology , Beclin-1/biosynthesis , Brain/metabolism , Microtubule-Associated Proteins/biosynthesis , Sequestosome-1 Protein/biosynthesis , Stroke/metabolism , Aged , Aged, 80 and over , Beclin-1/analysis , Brain/pathology , Brain Chemistry/physiology , Female , Humans , Male , Microtubule-Associated Proteins/analysis , Sequestosome-1 Protein/analysis , Stroke/pathology
12.
Exp Neurol ; 279: 86-95, 2016 May.
Article in English | MEDLINE | ID: mdl-26896832

ABSTRACT

Inhibition of the Rho/Rho kinase pathway has been shown to be beneficial in a variety of neural injuries and diseases. In this manuscript we investigate the role of Rho kinase inhibition in recovery from traumatic brain injury using a controlled cortical impact model in mice. Mice subjected to a moderately severe TBI were treated for 1 or 4 weeks with the Rho kinase inhibitor Y27632, and functional outcomes and neuronal and glial cell responses were analysed at 1, 7 and 35 days post-injury. We hypothesised that Y27632-treated mice would show functional improvement, with augmented recruitment of neuroblasts from the SVZ and enhanced survival of newborn neurons in the pericontusional cortex, with protection against neuronal degeneration, neuroinflammation and modulation of astrocyte reactivity and blood-brain-barrier permeability. While Rho kinase inhibition enhanced recovery of motor function after trauma, there were no substantial increases in the recruitment of DCX(+) neuroblasts or the number of BrdU(+) or EdU(+) labelled newborn neurons in the pericontusional cortex of Y27632-treated mice. Inhibition of Rho kinase significantly reduced the number of degenerating cortical neurons at 1day post-injury compared to saline controls but had no longer term effect on neuronal degeneration, with only modest effects on astrocytic reactivity and macrophage/microglial responses. Overall, this study showed that Rho kinase contributes to acute neurodegenerative processes in the injured cortex but does not play a significant role in SVZ neural precursor cell-derived adult neurogenesis, glial responses or blood-brain barrier permeability following a moderately severe brain injury.


Subject(s)
Amides/therapeutic use , Brain Injuries/drug therapy , Cell Survival/drug effects , Neuritis/drug therapy , Neurogenesis/drug effects , Neuroglia/drug effects , Neurons/drug effects , Neuroprotective Agents/therapeutic use , Protein Kinase Inhibitors/therapeutic use , Pyridines/therapeutic use , rho-Associated Kinases/antagonists & inhibitors , Amides/pharmacology , Animals , Astrocytes/drug effects , Blood-Brain Barrier/drug effects , Brain Injuries/pathology , Brain Injuries/psychology , Doublecortin Protein , Male , Mice , Mice, Inbred C57BL , Neuritis/pathology , Neuroprotective Agents/pharmacology , Protein Kinase Inhibitors/pharmacology , Psychomotor Performance/drug effects , Pyridines/pharmacology , Recovery of Function/drug effects , Treatment Outcome
13.
PLoS One ; 10(3): e0121541, 2015.
Article in English | MEDLINE | ID: mdl-25798924

ABSTRACT

Oligodendrocytes are responsible for producing and maintaining myelin throughout the CNS. One of the pathological features observed following traumatic brain injury (TBI) is the progressive demyelination and degeneration of axons within white matter tracts. While the effect of TBI on axonal health has been well documented, there is limited information regarding the response of oligodendrocytes within these areas. The aim of this study was to characterize the response of both mature oligodendrocytes and immature proliferative oligodendrocyte lineage cells across a 3 month timecourse following TBI. A computer-controlled cortical impact model was used to produce a focal lesion in the left motor cortex of adult mice. Immunohistochemical analyses were performed at 48 hours, 7 days, 2 weeks, 5 weeks and 3 months following injury to assess the prevalence of mature CC-1+ oligodendrocyte cell death, immature Olig2+ cell proliferation and longer term survival in the corpus callosum and external capsule. Decreased CC-1 immunoreactivity was observed in white matter adjacent to the site of injury from 2 days to 2 weeks post TBI, with ongoing mature oligodendrocyte apoptosis after this time. Conversely, proliferation of Olig2+ cells was observed as early as 48 hours post TBI and significant numbers of these cells and their progeny survived and remained in the external capsule within the injured hemisphere until at least 3 months post injury. These findings demonstrate that immature oligodendrocyte lineage cells respond to TBI by replacing oligodendrocytes lost due to damage and that this process occurs for months after injury.


Subject(s)
Brain Injuries/pathology , Oligodendroglia/pathology , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Death , Cell Lineage , Female , Male , Mice , Mice, Inbred C57BL , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Neural Stem Cells/metabolism , Neural Stem Cells/pathology , Oligodendrocyte Transcription Factor 2 , Oligodendroglia/metabolism
14.
Langmuir ; 30(29): 8898-906, 2014 Jul 29.
Article in English | MEDLINE | ID: mdl-24979524

ABSTRACT

The purpose of this work was to synthesize and screen, for their effectiveness to act as T1-enhancing magnetic resonance imaging (MRI) contrast agents, a small library of nitroxide lipids incorporated into cubic-phase lipid nanoparticles (cubosomes). The most effective nitroxide lipid was then formulated into lower-toxicity lipid nanoparticles (hexosomes), and effective MR contrast was observed in the aorta and spleen of live rats in vivo. This new class of lower-toxicity lipid nanoparticles allowed for higher relaxivities on the order of those of clinically used gadolinium complexes. The new hexosome formulation presented herein was significantly lower in toxicity and higher in relaxivity than cubosome formulations previously reported by us.


Subject(s)
Contrast Media/chemical synthesis , Magnetic Resonance Imaging/methods , Myristates/chemistry , Nanoparticles/chemistry , Nitrogen Oxides/chemistry , Animals , Aorta/anatomy & histology , CHO Cells , Cell Line, Tumor , Cell Survival/drug effects , Cricetulus , Erythrocytes/drug effects , Fatty Alcohols/chemistry , Female , Glycerides/chemistry , Humans , Mice , Mice, Inbred C57BL , Nanoparticles/ultrastructure , Rats , Rats, Sprague-Dawley , Spleen/anatomy & histology
15.
Cerebellum Ataxias ; 1: 18, 2014.
Article in English | MEDLINE | ID: mdl-26331042

ABSTRACT

BACKGROUND: Most adult-onset sporadic ataxias are unexplained, and the claim that many of these may be a result of gluten sensitivity has led to uncertainty as to whether to test for anti-gliadin antibodies (αGAb) and, if present, whether to recommend a gluten-free diet or continue searching for other causes of ataxia. This uncertainty arises in part from the frequency of αGAb in the population (about 1 in 10), but recent work delineating transglutaminase 6 as the target antigen in gluten ataxia has clarified the situation somewhat. Our aim was to determine whether there is molecular mimicry between cerebellar Purkinje cell antigens and gluten in subjects selected for recent diagnosis of CD rather than for ataxia. RESULTS: High titre αGAb sera from 11 newly-diagnosed CD patients and normal sera from 10 healthy controls were used to detect cross-reacting antibodies to cerebellar and cerebral cortex antigens in mouse, monkey and human tissue. None of the CD patients displayed ataxia. Mouse and human cerebellar and cerebral cortex extracts were analysed by Western blot probed with CD and control sera. Immunofluorescence microscopy was used on mouse and monkey cerebellar sections immunostained with CD and control sera to detect cross-reacting IgG antibodies. Western blot analysis of cerebellar and cerebral cortex extracts probed with CD sera did not demonstrate any specific immunoreactivity unique to the cerebellum. An identical twin pair with CD produced different patterns of reactivity. Immunofluorescence staining of mouse and monkey cerebellar sections showed most control and CD sera reacted non-specifically, with the exception of two CD and one control sera, each having a unique staining pattern. CONCLUSIONS: CD patient sera with high titre αGAb do not detect a common Purkinje cell or cerebellar-specific epitope. The pattern of reactivity is not solely dependent on genetic background.

16.
J Neurotrauma ; 31(7): 618-29, 2014 Apr 01.
Article in English | MEDLINE | ID: mdl-24279428

ABSTRACT

Secondary hypoxia is a known contributor to adverse outcomes in patients with traumatic brain injury (TBI). Based on the evidence that hypoxia and TBI in isolation induce neuroinflammation, we investigated whether TBI combined with hypoxia enhances cerebral cytokine production. We also explored whether increased concentrations of injury biomarkers discriminate between hypoxic (Hx) and normoxic (Nx) patients, correlate to worse outcome, and depend on blood-brain barrier (BBB) dysfunction. Forty-two TBI patients with Glasgow Coma Scale ≤8 were recruited. Cerebrospinal fluid (CSF) and serum were collected over 6 days. Patients were divided into Hx (n=22) and Nx (n=20) groups. Eight cytokines were measured in the CSF; albumin, S100, myelin basic protein (MBP) and neuronal specific enolase (NSE) were quantified in serum. CSF/serum albumin quotient was calculated for BBB function. Glasgow Outcome Scale Extended (GOSE) was assessed at 6 months post-TBI. Production of granulocye macrophage-colony stimulating factor (GM-CSF) was higher, and profiles of GM-CSF, interferon (IFN)-γ and, to a lesser extent, tumor necrosis factor (TNF), were prolonged in the CSF of Hx but not Nx patients at 4-5 days post-TBI. Interleukin (IL)-2, IL-4, IL-6, and IL-10 increased similarly in both Hx and Nx groups. S100, MBP, and NSE were significantly higher in Hx patients with unfavorable outcome. Among these three biomarkers, S100 showed the strongest correlations to GOSE after TBI-Hx. Elevated CSF/serum albumin quotients lasted for 5 days post-TBI and displayed similar profiles in Hx and Nx patients. We demonstrate for the first time that post-TBI hypoxia is associated with prolonged neuroinflammation, amplified extravasation of biomarkers, and poor outcome. S100 and MBP could be implemented to track the occurrence of post-TBI hypoxia, and prompt adequate treatment.


Subject(s)
Brain Injuries/physiopathology , Cytokines/biosynthesis , Hypoxia, Brain/physiopathology , Recovery of Function , Adolescent , Adult , Biomarkers/analysis , Blood-Brain Barrier/pathology , Brain Injuries/complications , Cytokines/analysis , Enzyme-Linked Immunosorbent Assay , Female , Glasgow Coma Scale , Humans , Hypoxia, Brain/complications , Male , Middle Aged , Prognosis , Young Adult
17.
Exp Neurol ; 252: 37-46, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24275527

ABSTRACT

There is controversy whether accumulation of the tumor suppressor PTEN protein in the cell nucleus under stress conditions such as trauma and stroke causes cell death. A number of in vitro studies have reported enhanced apoptosis in neurons possessing nuclear PTEN, with the interpretation that its nuclear phosphatase activity leads to reduction of the survival protein phospho-Akt. However, there have been no in vivo studies to show that nuclear PTEN in neurons under stress is detrimental. Using a mouse model of injury, we demonstrate here that brain trauma altered the nucleo-cytoplasmic distribution of Pten, resulting in increased nuclear Pten but only in surviving neurons near the lesion. This event was driven by Ndfip1, an adaptor and activator of protein ubiquitination by Nedd4 E3 ligases. Neurons next to the lesion with nuclear PTEN were invariably negative for TUNEL, a marker for cell death. These neurons also showed increased Ndfip1 which we previously showed to be associated with neuron survival. Biochemical assays revealed that overall levels of Pten in the affected cortex were unchanged after trauma, suggesting that Pten abundance globally had not increased but rather Pten subcellular location in affected neurons had changed. Following experimental injury, the number of neurons with nuclear Pten was reduced in heterozygous mice (Ndfip1(+/-)) although lesion volumes were increased. We conclude that nuclear trafficking of Pten following injury leads to neuron survival not death.


Subject(s)
Brain Injuries/pathology , Cell Nucleus/metabolism , Gene Expression Regulation/physiology , Neurons , PTEN Phosphohydrolase/metabolism , Analysis of Variance , Animals , Apoptosis/drug effects , Apoptosis/genetics , Apoptosis/physiology , Carrier Proteins/genetics , Cell Survival/genetics , Cytoplasm , Disease Models, Animal , Functional Laterality , Immunoprecipitation , In Situ Nick-End Labeling , Intercellular Signaling Peptides and Proteins , Membrane Proteins/deficiency , Membrane Proteins/genetics , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neurons/metabolism , Neurons/pathology , Neurons/ultrastructure , Oncogene Protein v-akt , PTEN Phosphohydrolase/genetics , Protein Transport/genetics
18.
J Neuroinflammation ; 10: 156, 2013 Dec 18.
Article in English | MEDLINE | ID: mdl-24344874

ABSTRACT

BACKGROUND: Diffuse axonal injury is a common consequence of traumatic brain injury (TBI) and often co-occurs with hypoxia, resulting in poor neurological outcome for which there is no current therapy. Here, we investigate the ability of the multifunctional compound erythropoietin (EPO) to provide neuroprotection when administered to rats after diffuse TBI alone or with post-traumatic hypoxia. METHODS: Sprague-Dawley rats were subjected to diffuse traumatic axonal injury (TAI) followed by 30 minutes of hypoxic (Hx, 12% O2) or normoxic ventilation, and were administered recombinant human EPO-α (5000 IU/kg) or saline at 1 and 24 hours post-injury. The parameters examined included: 1) behavioural and cognitive deficit using the Rotarod, open field and novel object recognition tests; 2) axonal pathology (NF-200); 3) callosal degradation (hematoxylin and eosin stain); 3) dendritic loss (MAP2); 4) expression and localisation of the EPO receptor (EpoR); 5) activation/infiltration of microglia/macrophages (CD68) and production of IL-1ß. RESULTS: EPO significantly improved sensorimotor and cognitive recovery when administered to TAI rats with hypoxia (TAI + Hx). A single dose of EPO at 1 hour reduced axonal damage in the white matter of TAI + Hx rats at 1 day by 60% compared to vehicle. MAP2 was decreased in the lateral septal nucleus of TAI + Hx rats; however, EPO prevented this loss, and maintained MAP2 density over time. EPO administration elicited an early enhanced expression of EpoR 1 day after TAI + Hx compared with a 7-day peak in vehicle controls. Furthermore, EPO reduced IL-1ß to sham levels 2 hours after TAI + Hx, concomitant to a decrease in CD68 positive cells at 7 and 14 days. CONCLUSIONS: When administered EPO, TAI + Hx rats had improved behavioural and cognitive performance, attenuated white matter damage, resolution of neuronal damage spanning from the axon to the dendrite, and suppressed neuroinflammation, alongside enhanced expression of EpoR. These data provide compelling evidence of EPO's neuroprotective capability. Few benefits were observed when EPO was administered to TAI rats without hypoxia, indicating that EPO's neuroprotective capacity is bolstered under hypoxic conditions, which may be an important consideration when EPO is employed for neuroprotection in the clinic.


Subject(s)
Brain Injuries/pathology , Erythropoietin/pharmacology , Neuroprotective Agents/pharmacology , Recovery of Function/drug effects , Animals , Axons/drug effects , Axons/pathology , Behavior, Animal/drug effects , Brain Injuries/metabolism , Hypoxia, Brain/metabolism , Hypoxia, Brain/pathology , Immunohistochemistry , Inflammation/pathology , Male , Motor Activity/drug effects , Rats , Rats, Sprague-Dawley , Receptors, Erythropoietin/metabolism , Up-Regulation
19.
Neurosignals ; 20(3): 132-46, 2012.
Article in English | MEDLINE | ID: mdl-22456466

ABSTRACT

Brain injury following stroke or trauma induces the migration of neuroblasts derived from subventricular zone neural precursor cells (NPCs) towards the damaged tissue, where they then have the potential to contribute to repair. Enhancing the recruitment of new cells thus presents an enticing prospect for the development of new therapeutic approaches to treat brain injury; to this end, an understanding of the factors regulating this process is required. During the neuroinflammatory response to ischemic and traumatic brain injuries, a plethora of pro- and anti-inflammatory cytokines, chemokines and growth factors are released in the damaged tissue, and recent work indicates that a variety of these are able to influence injury-induced migration. In this review, we will discuss the contribution of specific chemokines and growth factors towards stimulating NPC migration in the injured brain.


Subject(s)
Brain Injuries/pathology , Brain Ischemia/pathology , Cell Movement/physiology , Inflammation/pathology , Neurons/cytology , Animals , Brain Injuries/physiopathology , Brain Ischemia/physiopathology , Inflammation/physiopathology , Neurogenesis/physiology , Neurons/physiology
20.
J Neurotrauma ; 29(7): 1410-25, 2012 May 01.
Article in English | MEDLINE | ID: mdl-22260446

ABSTRACT

Neurogenesis is stimulated following injury to the adult brain and could potentially contribute to tissue repair. However, evidence suggests that microglia activated in response to injury are detrimental to the survival of new neurons, thus limiting the neurogenic response. The aim of this study was to determine the effect of the anti-inflammatory drug minocycline on neurogenesis and functional recovery after a closed head injury model of focal traumatic brain injury (TBI). Beginning 30 min after trauma, minocycline was administered for up to 2 weeks and bromodeoxyuridine was given on days 1-4 to label proliferating cells. Neurological outcome and motor function were evaluated over 6 weeks using the Neurological Severity Score (NSS) and ledged beam task. Microglial activation was assessed in the pericontusional cortex and hippocampus at 1 week post-trauma, using immunohistochemistry to detect F4/80. Following immunolabeling of bromodeoxyuridine, double-cortin, and NeuN, cells undergoing distinct stages of neurogenesis, including proliferation, neuronal differentiation, neuroblast migration, and long-term survival, were quantified at 1 and 6 weeks in the hippocampal dentate gyrus, as well as in the subventricular zone of the lateral ventricles and the pericontusional cortex. Our results show that minocycline successfully reduced microglial activation and promoted early neurological recovery that was sustained over 6 weeks. We also show for the first time in the closed head injury model, that early stages of neurogenesis were stimulated in the hippocampus and subventricular zone; however, no increase in new mature neurons occurred. Contrary to our hypothesis, despite the attenuation of activated microglia, minocycline did not support neurogenesis in the hippocampus, lateral ventricles, or pericontusional cortex, with none of the neurogenic stages being affected by treatment. These data provide evidence that a general suppression of microglial activation is insufficient to enhance neuronal production, suggesting that further work is required to elucidate the relationship between microglia and neurogenesis after TBI.


Subject(s)
Brain Injuries/pathology , Microglia/metabolism , Microglia/pathology , Minocycline/pharmacology , Neurogenesis/drug effects , Animals , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Brain Injuries/drug therapy , Cell Proliferation/drug effects , Disease Models, Animal , Mice , Mice, Inbred C57BL , Microglia/drug effects , Minocycline/therapeutic use , Neurogenesis/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...