9 research outputs found

    A tale of two towns: A comparative study exploring the possibilities and pitfalls of social capital among people seeking recovery from substance misuse

    Get PDF
    Background: Social capital has become an influential concept in debating and understanding the modern world. Within the drug and alcohol sector, the concept of ‘recovery capital’ has gained traction with researchers suggesting that people who have access to such capital are better placed to overcome their substance use-related problems than those who do not (Cloud and Granfield, 2008), leading to requests for interventions that focus on building social capital networks (Neale & Stevenson, 2015). While accepting that the concept of social capital has enormous potential for addressing the problems associated with drug use, this paper also considers its ‘dark side’. Methods: Data were drawn from semi-structured interviews with 180 participants including 135 people who use drugs and 45 people who formerly used drugs. Results: High levels of trust, acquired through the establishment of dense social networks, are required to initiate recovery. However, these ‘strong bonds’ may also lead to the emergence of what is perceived by others as an exclusive social network that limits membership to those who qualify and abide by the ‘rules’ of the recovery community, particularly around continuous abstinence. Conclusions: Depending on the nature of the networks and the types of links participants have into them being socially connected can both inhibit and encourage recovery. Therefore, the successful application of social capital within the drugs and alcohol field requires a consideration of not only the presence or absence of social connections but their nature, the value they produce, and the social contexts within which they are developed

    Using light sheet microscopy to investigate the role of apolipoprotein e4 in traumatic vascular injury

    No full text
    Neurovascular impairments such as blood brain barrier (BBB) dysfunction, decreased cerebral blood flow, and small vessel disease have been associated with traumatic brain injury (TBI), dementia and other neurological disorders, and contribute to cognitive impairment. Due to the complex structure of the cerebral vasculature, it is technically challenging to routinely analyse the murine vasculature at high resolution using traditional histopathological or magnetic resonance imaging (MRI) approaches. The objective of this thesis is to develop an experimental pipeline to map the cerebral vasculature using tissue clearing, light sheet microscopy (LSM) and 3D image analysis to provide high-resolution characterization of the structure and integrity of the entire murine vasculature after TBI. Additionally, Apolipoprotein E (ApoE), which carries lipids in the brain in the form of lipoproteins, is the major genetic risk factor for sporadic Alzheimer’s Disease (AD). Recent studies have shown that APOE can exacerbate AD pathology via vascular pathways, and even in the absence of AD diagnosis, the APOE4 allele has emerged as a risk factor for small vessel disease and vascular cognitive impairment in comparison to the more common isoform APOE3. As proof-of-concept of the utility of our 3D analysis pipeline, we used the CHIMERA (Closed-Head Impact Model of Engineered Rotational Acceleration) TBI model to subject both ApoE4 targeted replacement (ApoE4-TR) and ApoE3 targeted replacement (ApoE3-TR) mice to TBI or sham procedures followed by transcardial perfusion of florescent wheat germ agglutinin (WGA) to label the whole brain vasculature. Brains were fixed using a technique called stabilization under harsh conditions via intramolecular epoxide linkages to prevent degradation (SHIELD) and passively cleared for imaging of WGA-labelled vessels using LSM. Volumetric images were stitched, 3D rendered and analyzed using Imaris image analysis software, to achieve a resolution of 1250x1250 pixels with an image resolution at pixel size of 1.5 x 1.5 x 5 µm (xyz plane). In summary, the work presented here provides proof of concept data of the establishment of an efficient imaging and analysis pipeline, which has great flexibility to be applied to a myriad of research projects examining different pathologies of the murine brain.Medicine, Faculty ofPathology and Laboratory Medicine, Department ofGraduat

    Altered Tau Kinase Activity in rTg4510 Mice after a Single Interfaced CHIMERA Traumatic Brain Injury

    No full text
    Traumatic brain injury (TBI) is an established risk factor for neurodegenerative diseases. In this study, we used the Closed Head Injury Model of Engineered Rotational Acceleration (CHIMERA) to investigate the effects of a single high-energy TBI in rTg4510 mice, a mouse model of tauopathy. Fifteen male rTg4510 mice (4 mo) were impacted at 4.0 J using interfaced CHIMERA and were compared to sham controls. Immediately after injury, the TBI mice showed significant mortality (7/15; 47%) and a prolonged duration of loss of the righting reflex. At 2 mo post-injury, surviving mice displayed significant microgliosis (Iba1) and axonal injury (Neurosilver). Western blotting indicated a reduced p-GSK-3β (S9):GSK-3β ratio in TBI mice, suggesting chronic activation of tau kinase. Although longitudinal analysis of plasma total tau suggested that TBI accelerates the appearance of tau in the circulation, there were no significant differences in brain total or p-tau levels, nor did we observe evidence of enhanced neurodegeneration in TBI mice compared to sham mice. In summary, we showed that a single high-energy head impact induces chronic white matter injury and altered GSK-3β activity without an apparent change in post-injury tauopathy in rTg4510 mice.Applied Science, Faculty ofMedicine, Faculty ofOther UBCNon UBCBiomedical Engineering, School ofPathology and Laboratory Medicine, Department ofReviewedFacultyResearcherPostdoctoralGraduat

    Development of a novel, sensitive translational immunoassay to detect plasma glial fibrillary acidic protein (GFAP) after murine traumatic brain injury

    No full text
    Background: Glial fibrillary acidic protein (GFAP) has emerged as a promising fluid biomarker for several neurological indications including traumatic brain injury (TBI), a leading cause of death and disability worldwide. In humans, serum or plasma GFAP levels can predict brain abnormalities including hemorrhage on computed tomography (CT) scans and magnetic resonance imaging (MRI). However, assays to quantify plasma or serum GFAP in preclinical models are not yet available. Methods: We developed and validated a novel sensitive GFAP immunoassay assay for mouse plasma on the Meso Scale Discovery immunoassay platform and validated assay performance for robustness, precision, limits of quantification, dilutional linearity, parallelism, recovery, stability, selectivity, and pre-analytical factors. To provide proof-of-concept data for this assay as a translational research tool for TBI and Alzheimer’s disease (AD), plasma GFAP was measured in mice exposed to TBI using the Closed Head Impact Model of Engineered Rotational Acceleration (CHIMERA) model and in APP/PS1 mice with normal or reduced levels of plasma high-density lipoprotein (HDL). Results: We performed a partial validation of our novel assay and found its performance by the parameters studied was similar to assays used to quantify human GFAP in clinical neurotrauma blood specimens and to assays used to measure murine GFAP in tissues. Specifically, we demonstrated an intra-assay CV of 5.0%, an inter-assay CV of 7.2%, a lower limit of detection (LLOD) of 9.0 pg/mL, a lower limit of quantification (LLOQ) of 24.8 pg/mL, an upper limit of quantification (ULOQ) of at least 16,533.9 pg/mL, dilution linearity of calibrators from 20 to 200,000 pg/mL with 90–123% recovery, dilution linearity of plasma specimens up to 32-fold with 96–112% recovery, spike recovery of 67–100%, and excellent analyte stability in specimens exposed to up to 7 freeze-thaw cycles, 168 h at 4 °C, 24 h at room temperature (RT), or 30 days at − 20 °C. We also observed elevated plasma GFAP in mice 6 h after TBI and in aged APP/PS1 mice with plasma HDL deficiency. This assay also detects GFAP in serum. Conclusions: This novel assay is a valuable translational tool that may help to provide insights into the mechanistic pathophysiology of TBI and AD.Applied Science, Faculty ofMedicine, Faculty ofOther UBCBiomedical Engineering, School ofPathology and Laboratory Medicine, Department ofReviewedFacult

    CHIMERA repetitive mild traumatic brain injury induces chronic behavioural and neuropathological phenotypes in wild-type and APP/PS1 mice

    No full text
    Background: The annual incidence of traumatic brain injury (TBI) in the United States is over 2.5 million, with approximately 3–5 million people living with chronic sequelae. Compared with moderate-severe TBI, the long-term effects of mild TBI (mTBI) are less understood but important to address, particularly for contact sport athletes and military personnel who have high mTBI exposure. The purpose of this study was to determine the behavioural and neuropathological phenotypes induced by the Closed-Head Impact Model of Engineered Rotational Acceleration (CHIMERA) model of mTBI in both wild-type (WT) and APP/PS1 mice up to 8 months post-injury. Methods: Male WT and APP/PS1 littermates were randomized to sham or repetitive mild TBI (rmTBI; 2 × 0.5 J impacts 24 h apart) groups at 5.7 months of age. Animals were assessed up to 8 months post-injury for acute neurological deficits using the loss of righting reflex (LRR) and Neurological Severity Score (NSS) tasks, and chronic behavioural changes using the passive avoidance (PA), Barnes maze (BM), elevated plus maze (EPM) and rotarod (RR) tasks. Neuropathological assessments included white matter damage; grey matter inflammation; and measures of Aβ levels, deposition, and aducanumab binding activity. Results: The very mild CHIMERA rmTBI conditions used here produced no significant acute neurological or motor deficits in WT and APP/PS1 mice, but they profoundly inhibited extinction of fear memory specifically in APP/PS1 mice over the 8-month assessment period. Spatial learning and memory were affected by both injury and genotype. Anxiety and risk-taking behaviour were affected by injury but not genotype. CHIMERA rmTBI induced chronic white matter microgliosis, axonal injury and astrogliosis independent of genotype in the optic tract but not the corpus callosum, and it altered microgliosis in APP/PS1 amygdala and hippocampus. Finally, rmTBI did not alter long-term tau, Aβ or amyloid levels, but it increased aducanumab binding activity. Conclusions: CHIMERA is a useful model to investigate the chronic consequences of rmTBI, including behavioural abnormalities consistent with features of post-traumatic stress disorder and inflammation of both white and grey matter. The presence of human Aβ greatly modified extinction of fear memory after rmTBI.Applied Science, Faculty ofMedicine, Faculty ofOther UBCMechanical Engineering, Department ofMedicine, Department ofNeurology, Division ofPathology and Laboratory Medicine, Department ofReviewedFacult

    Property as Sequential Exchange: The Forgotten Limits of Private Contract

    No full text
    corecore